Purinergic Receptors and their Modulators (Topics in Medicinal Chemistry, 41) 303139724X, 9783031397240

This book overviews purinergic receptors that are playing key roles in human and pathophysiological processes. The book

123 26 10MB

English Pages 364 [356] Year 2023

Report DMCA / Copyright

DOWNLOAD PDF FILE

Table of contents :
Preface
Contents
Once Upon a Time Adenosine and Its Receptors: Historical Survey and Perspectives as Potential Targets for Therapy in Human Dis...
1 Introduction
2 Brief History of Adenosine and Its Receptors
3 The State of the Art
3.1 Adenosine Production and Metabolism
3.2 Distribution of Adenosine Receptors and Their Main Physiological Effects
3.3 Signal Transduction of Adenosine Receptors
3.4 Adenosine Receptor Structures
3.5 Medicinal Chemistry of Adenosine Receptor
3.5.1 Adenosine Receptor Ligands, in Short
3.5.2 Emerging Adenosine Receptor Ligands as Tool Compounds
Agonists
Antagonists
3.6 Pathophysiological Roles of Adenosine Receptors and Emerging Therapeutic Intervention
3.6.1 Central Nervous System Disorders
3.6.2 Pain
3.6.3 Inflammation and Immune Diseases
3.6.4 Cancer
3.6.5 Cardiovascular and Related Diseases
4 Perspectives
5 Concluding Remarks
References
Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year History of Pyrazolo[4,3-e][1,2,4]Triazolo[1,5-c]Pyrim...
1 Introduction
1.1 A1 AR
1.2 A2A AR
1.3 A2B AR
1.4 A3 AR
2 Pyrazolo-Triazolo-Pyrimidines (PTP) as AR Antagonists
2.1 A2A AR Antagonists
2.2 A3 AR Antagonists
2.3 A2B and A1 ARs Antagonists
3 Probes and Functional Conjugates Targeting Adenosine Receptors Bearing a PTP Scaffold
3.1 Covalent Derivatives
3.2 Radiolabeled Derivatives
3.3 Conjugated Derivatives
3.3.1 Fluorescent Derivatives
3.3.2 Other Conjugates
4 Conclusions
References
Adenosine Receptor Ligands as Potential Therapeutic Agents for Impaired Wound Healing and Fibrosis
1 Introduction
2 The Wound Healing Process
3 Adenosine in Wound Healing
4 Adenosine and Fibrosis
5 Conclusions
References
Adenosine A2A Receptor Antagonists: Chemistry, SARs, and Therapeutic Potential
1 Introduction
2 A2A Adenosine Receptor Antagonists Evaluated in Clinical Trials
3 A2A Adenosine Receptor Antagonists
3.1 Xanthine Derivatives
3.2 Non-Xanthine Derivatives
3.2.1 Triazolo-Quinazoline and -Quinoxaline Derivatives
3.2.2 Pyrazolo-, Imidazolo-, and Thiazolo-Triazolopyrimidine Derivatives
3.2.3 Triazolo-Triazine, -Pyrimidine, and -Pyrazine Derivatives
3.2.4 Thiazolo- and Thieno-Pyrimidine Derivatives
3.2.5 Benzo-, Pyrimido-, and Oxazolo-Thiazolo Derivatives
3.2.6 Purine and Adenine Derivatives
3.2.7 Thiazole and Triazole Derivatives
3.2.8 Pyridine and Pyrimidine Derivatives
3.2.9 Pyrazine and Triazine Derivatives
4 Structural Features of A2A Structure and Its Interaction with Antagonists
5 Potential Application of A2A Adenosine Receptor Antagonists
5.1 A2A Adenosine Receptor Antagonists and Neurodegenerative Disorders
5.2 A2A Adenosine Receptor Antagonists and Neuroprotection
5.3 A2A Adenosine Receptor Antagonists and Cancer
6 Concluding Remarks
References
A2B Adenosine Receptor as a New and Attractive Target to Treat Brain Ischemia or Demyelination
1 Introduction
2 Role of A2BRs in Cerebral Ischemia
3 Role of A2BRs in Demyelinating Conditions
4 Conclusions
References
A3 Adenosine Receptor Ligands: From Discovery to Clinical Trials
1 Introduction
1.1 Historical Sketch: A3AR Discovery
2 A3AR Agonists
2.1 Early Medicinal Chemistry of A3AR Agonists
2.2 Recently Reported A3AR Agonists
3 A3AR Antagonists
3.1 Early Medicinal Chemistry of A3AR Antagonists
3.2 Recently Reported A3AR Antagonists
4 A3AR Prodrugs
5 A3AR Allosteric Modulators
6 A3AR Molecular Modeling
7 Clinical Trials of A3AR Ligands
8 Conclusions
References
Bifunctional Tools to Study Adenosine Receptors
1 Introduction
1.1 General Design Aspects of Bifunctional Adenosine Receptor Tools
2 Heterobivalent Ligands
2.1 Heterobivalent A1R Ligands
2.2 Heterobivalent A2AR Ligands
2.3 Heterobivalent Ligands Targeting A3R
2.4 Structural Biology and Molecular Modelling of Bivalent Ligands
3 Dual-Acting Ligands
3.1 Dual-Acting Ligands for Neurodegenerative Diseases/Conditions
3.2 Dual-Acting Ligands for Anticancer, Cardioprotective, Anti-Diabetic and Other Applications
3.3 Dual-Acting Prodrugs
4 Bitopic Ligands
4.1 Bitopic A1R Ligands
4.2 Bitopic A2AR, A2BR and A3R Ligands
5 Conclusions
References
Allosteric Modulators of Adenosine Receptors
1 Introduction
2 Evolution of the Allosteric Models
3 Allosteric Modulation of GPCR
3.1 Methodological Aspect for Allosteric Ligand Characterization
3.2 GPCR Bias Allosteric Modulators (BAMs)
3.3 Advantages and Disadvantages of GPCR Allosteric Modulators
4 Mechanism of Adenosine Receptor (ARs) Allostery
4.1 AR Homo- and Hetero-Dimerization as an Allosteric Mechanism
4.2 Allosteric Modulation of AR by Sodium and Cholesterol
5 Allosteric Modulators of Adenosine Receptors
5.1 A1 AR Allosteric Modulators
5.1.1 Bitopic Allosteric Modulators of the A1AR
5.1.2 Biological Activity
5.2 A2A Allosteric Modulators
5.2.1 Biological Activity
5.3 A2B AR Allosteric Modulators
5.3.1 Biological Activity
5.4 A3 AR Allosteric Modulators
5.4.1 Biological Activity
6 Final Remarks
References
In Silico Insights Toward the Exploration of Adenosine Receptors Ligand Recognition
1 The Drug Discovery Process
2 The Benefits of Computation and Automation
3 The Main Branches in Computer-Aided Drug Design
4 Structural Biology Elements in Adenosine Receptor Research
5 Structure-Based Drug Design (SBDD)
5.1 Molecular Docking
5.2 Molecular Dynamics
5.3 Post-Docking Refinement
5.4 Enhanced Sampling Approaches
5.5 Free-Energy Perturbation (FEP)
6 Ligand-Based Drug Design (LBDD)
7 Machine Learning and Artificial Intelligence in Drug Discovery
7.1 The Machine Learning Method
7.2 Machine Learning in Drug Discovery and Its Implementation in the Adenosine Receptor Field
8 Conclusions
References
P2X4 Receptors in Immunity and Inflammation
1 Introduction
2 Purinergic Signaling: Receptors, Enzymes, Transporters
3 Structure and Molecular Biology of P2X4 Receptor
4 P2X4 Receptor and in Innate and Adaptive Immunity
4.1 P2X4 and Innate Immune Cells
4.1.1 Monocytes and Macrophages
4.1.2 Microglia
4.1.3 Dendritic Cells
4.1.4 Neutrophils
4.2 P2X4 and Adaptive Immune Cells
4.2.1 T Cells
4.2.2 B Cell
References
CD73 Inhibitors as Antitumor Agents
1 Introduction
2 CD73 Structure and Functions
2.1 CD73 Structure
2.2 CD73 Catalytic Mechanism
3 Nucleoside/Nucleotide and Non-nucleotide CD73 Inhibitors
3.1 Nucleoside/Nucleotide CD73 Inhibitors
3.2 Non-nucleotide Inhibitors
3.2.1 Monoclonal Antibodies (MAbs) as hCD73 Inhibitors
4 Conclusions
References
Bacterial Ectonucleotidases: Underexplored Antibacterial Drug Targets
1 Introduction
2 The Physiological Role of the Bacterial 5′NTs
2.1 Intracellular 5′-NTs
2.2 Membrane-Bound and Periplasmic 5′-NTs
3 Bacterial 5′-NTs as Pharmacological Targets
3.1 5′-NTs and Bacterial Virulence
3.2 Bacterial 5′-NT Inhibitors
4 Conclusions
References
Recommend Papers

Purinergic Receptors and their Modulators (Topics in Medicinal Chemistry, 41)
 303139724X, 9783031397240

  • 0 0 0
  • Like this paper and download? You can publish your own PDF file online for free in a few minutes! Sign Up
File loading please wait...
Citation preview

Topics in Medicinal Chemistry  41

Vittoria Colotta Claudiu T. Supuran   Editors

Purinergic Receptors and their Modulators

Topics in Medicinal Chemistry Volume 41

Series Editors Peter R. Bernstein, Philadelphia, PA, USA Amanda L. Garner, Ann Arbor, MI, USA Gunda I. Georg, Minneapolis, MN, USA Stefan Laufer, Tübingen, Germany John A. Lowe, Stonington, CT, USA Nicholas A. Meanwell, Princeton, NJ, USA Anil Kumar Saxena, Kashipur, India Claudiu T. Supuran, Sesto Fiorentino, Italy Ao Zhang, Shanghai, China Nuska Tschammer, Martinsried, Germany Sally-Ann Poulsen, Nathan, Australia

Topics in Medicinal Chemistry (TMC) covers all relevant aspects of medicinal chemistry research, e.g. pathobiochemistry of diseases, identification and validation of (emerging) drug targets, structural biology, drugability of targets, drug design approaches, chemogenomics, synthetic chemistry including combinatorial methods, bioorganic chemistry, natural compounds, high-throughput screening, pharmacological in vitro and in vivo investigations, drug-receptor interactions on the molecular level, structure-activity relationships, drug absorption, distribution, metabolism, elimination, toxicology and pharmacogenomics. Drug research requires interdisciplinary team-work at the interface between chemistry, biology and medicine. To fulfil this need, TMC is intended for researchers and experts working in academia and in the pharmaceutical industry, and also for graduates that look for a carefully selected collection of high quality review articles on their respective field of expertise. Medicinal chemistry is both science and art. The science of medicinal chemistry offers mankind one of its best hopes for improving the quality of life. The art of medicinal chemistry continues to challenge its practitioners with the need for both intuition and experience to discover new drugs. Hence sharing the experience of drug research is uniquely beneficial to the field of medicinal chemistry. All chapters from Topics in Medicinal Chemistry are published OnlineFirst with an individual DOI. In references, Topics in Medicinal Chemistry is abbreviated as Top Med Chem and cited as a journal.

Vittoria Colotta • Claudiu T. Supuran Editors

Purinergic Receptors and their Modulators With contributions by L. Antonioli  J. K. Awalt  E. Barresi  D. Bassani  L. Benvenuti  M. Buccioni  S. Calenda  C. Capasso  D. Catarzi  E. Cescon  C. Chang  F. Cherchi  V. Colotta  E. Coppi  V. D’Antongiovanni  T. Da Ros  F. Da Settimo  D. Dal Ben  I. Dettori  C. Di Salvo  S. Federico  M. Fornai  B. Francucci  C. Giacomelli  I. Grieco  G. Haskó  K. A. Jacobson  M. Jörg  C. Lambertucci  C. Martini  G. Marucci  L. T. May  S. Moro  A. Nocentini  P. Oliva  C. Payne  F. Pedata  C. Pellegrini  M. Persico  F. Prencipe  A. M. Pugliese  G. Spalluto  A. Spinaci  C. T. Supuran  R. R. Suresh  S. Taliani  M. L. Trincavelli  J. D. A. Tyndall  F. Varano  M. Venturini  A. J. Vernall  E. Vigiani  R. Volpini

Editors Vittoria Colotta Department of Neurofarba, Pharmaceutical and Nutraceutical Section University of Florence Florence, Italy

Claudiu T. Supuran Department of Neurofarba, Pharmaceutical and Nutraceutical Section University of Florence Florence, Italy

ISSN 1862-2461 ISSN 1862-247X (electronic) Topics in Medicinal Chemistry ISBN 978-3-031-39724-0 ISBN 978-3-031-39725-7 (eBook) https://doi.org/10.1007/978-3-031-39725-7 © The Editor(s) (if applicable) and The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 This work is subject to copyright. All rights are solely and exclusively licensed by the Publisher, whether the whole or part of the material is concerned, specifically the rights of translation, reprinting, reuse of illustrations, recitation, broadcasting, reproduction on microfilms or in any other physical way, and transmission or information storage and retrieval, electronic adaptation, computer software, or by similar or dissimilar methodology now known or hereafter developed. The use of general descriptive names, registered names, trademarks, service marks, etc. in this publication does not imply, even in the absence of a specific statement, that such names are exempt from the relevant protective laws and regulations and therefore free for general use. The publisher, the authors, and the editors are safe to assume that the advice and information in this book are believed to be true and accurate at the date of publication. Neither the publisher nor the authors or the editors give a warranty, expressed or implied, with respect to the material contained herein or for any errors or omissions that may have been made. The publisher remains neutral with regard to jurisdictional claims in published maps and institutional affiliations. This Springer imprint is published by the registered company Springer Nature Switzerland AG The registered company address is: Gewerbestrasse 11, 6330 Cham, Switzerland Paper in this product is recyclable.

Preface

Purinergic receptors, termed P1 and P2, are activated, respectively, by adenosine and different nucleotides, such as ATP and ADP. Purinergic signaling plays a key role in human physiological and pathological processes and many selective P1 and P2 ligands have been developed as therapeutics for a variety of diseases, such as thrombosis, cerebral ischemia, dry eye, neurodegenerative diseases, and cancer. Several P1 and P2 receptor modulators entered clinical trials and some have been approved for marketing. In this volume of the book series Topics in Medicinal Chemistry, we propose a series of reviews that provide updated information on purinergic receptors, their modulators, and related therapeutic applications and also give suggestions for possible future direction of research in this area. The first chapter, by Catarzi et al., is a detailed overview of adenosine and its receptors that helps the reader to enter the intriguing world of this purine nucleoside. Adenosine is an autacoid that locally regulates several tissue functions and also acts as a neuromodulator. It exerts its effects by activating G protein-coupled receptors, termed P1 receptors, and classified into A1, A2A, A2B, and A3 subtypes. Under metabolically unfavorable conditions such as tissue hypoxia or stress, adenosine counteracts tissue damage, but its excessive and chronic production can be harmful, leading to fibrosis, inflammation, and other pathological situations. This evidence made soon consider adenosine receptors (ARs) as potential drug targets and stimulated medicinal chemistry studies to produce a large number of AR ligands. The chapter summarizes the most relevant series of selective agonists and antagonists of the four AR subtypes, with a particular emphasis on the compounds employed as pharmacological tools, and as radioligands. A section reviews the effects and usefulness of AR ligands in different therapeutic areas (central nervous system, cardiovascular, inflammatory, and immune system, pain). Particular attention has been paid to binding kinetics and biased signaling of ARs that should be considered in the design of new AR ligands as more effective and safer drugs. The second chapter, by Federico et al., focuses on the twenty-year history of one of the most fruitful and deeply investigated series of AR antagonists, the pyrazolo [4,3-e][1,2,4]triazolo[1,5-c]pyrimidines (PTP). This series furnished many potent v

vi

Preface

and selective antagonists for the A2A and A3 subtypes some of which have been used as molecular probes for the study of structure and function of the A2A and A3 ARs. Both these AR subtypes have aroused great interest in their role in Parkinson’s disease and immune-oncology (A2A) and in cancer, glaucoma, and inflammation (A3). Structure-activity relationships at all ARs are discussed, pointing out the structural requirements to bind selectively the A2A or the A3A AR subtypes. PTP derivatives have also been taken as lead to the design of new molecular probes and multifunctional molecules. Several PTP-fluorophore conjugates were developed obtaining fluorescent antagonists toward A2A and/or A3 ARs, whose applications range from binding assays to fluorescence microscopy and receptor detection. PTPbased molecules were also described as dual ligands blocking both A2AAR and histone deacetylase as new potential drugs in cancer immunotherapy. The third chapter, by Varano et al., addresses particular potential therapeutic applications of AR ligands, i.e. treatment of wound healing and fibrosis. Agonists of the A2A AR, such as CGS21680 and sonedenoson (MRE-0094), accelerated wound closure and healing in animal models by a mechanism that depends on tissue plasminogen activator and angiogenesis promotion. Excessive activation of ARs can lead to matrix overproduction, scarring, and fibrosis of the skin, and organs such as the liver, heart, lungs, and kidneys. Thus, AR antagonists could represent novel therapeutic agents for the treatment of fibrotic diseases. A2A AR antagonists proved to be potentially useful against dermal and hepatic fibrosis, and the A2B AR blockade potentially leads to pulmonary fibrosis control. The high therapeutic potential demonstrated by A2AAR antagonists has prompted medicinal chemists to design and develop several series of compounds belonging to different chemical classes. The fourth chapter, by Volpini et al., reviews these series, from the first alkyl xanthine compounds to the most potent and selective heterocyclic derivatives unrelated to xanthines. Structure–activity relationship studies are described for the most relevant classes. An overview of the A2AAR antagonists evaluated in clinical trials is provided and one section is dedicated to the representation of published crystallographic structures and molecular modeling studies. Furthermore, the use of A2A AR antagonists as neuroprotective agents and their potential application in the anti-cancer immune therapy is discussed. In contrast to the A2A AR, the A2B AR has been less investigated due to the scarce availability of truly selective agonists and antagonists of this AR subtype. The fifth chapter, by Cherchi et al, is an overview of recent knowledge acquired on the role of the A2B AR subtype in brain ischemia. The results of different research are critically discussed pointing out the diverse and contradictory roles played by this receptor after ischemia, being harmful or protective, depending on the timing and its cellular localization. The role of the A2BAR in demyelinating conditions, occurring after ischemic injury, has also been reviewed highlighting that the A2BAR antagonists improve remyelination and, consequently, white matter integrity in stroke and demyelinating pathologies. The A3AR receptor was the last of the four ARs to be discovered, it was identified by cloning and first described as an orphan receptor. The sixth chapter, by Jacobson et al., focuses on this AR subtype, dealing with the medicinal chemistry of its

Preface

vii

selective agonists, antagonists, and allosteric modulators, from the early discoveries to the most recent advances. Radiolabeled ligands developed as receptor probes are described, as well as prodrugs of some derivatives and their efficacy. A section reports on the grown number of clinical trials conducted on A3AR ligands which proved to be therapeutic candidate molecules for a variety of chronic (e.g., psoriasis, liver disease, pain, cancer, glaucoma) and acute (stroke, cardiac ischemia) conditions. Bifunctional compounds designed to target ARs and comprising bivalent, bitopic, and dual-acting ligands are reviewed in Chap. 7, by Vernall et al. The beginning of the chapter provides a clear description of the bifunctional ligands. In the last years, the interest in multi-target ligands has progressively increased due to their potential benefits for the treatment of complex diseases such as cancer and CNS disorders. The development of bivalent ligands is based on the widely accepted evidence that G protein-coupled receptors, including ARs, can exist as homodimers or heterodimers. Bivalent ligands targeting the heterodimers A1AR-β2R, A1AR-D1R, A2AAR-D2R, A3AR-A1AR are reported. Dual-acting ligands behaving as A1AR-A2AAR antagonists, A2AAR antagonists-MAO-B inhibitors, and A2A-D1 antagonists, all developed as potential anti-PD agents, have been inserted. A section is devoted to discussing the fundamental role of computational methods and structural biology in guiding the rational design of bifunctional ligands. Chapter 8, by Taliani et al., deals with allosteric modulators of ARs, the first GPCRs known to be allosterically regulated. The chapter begins with a detailed description of the main mechanisms of GPCRs’ allosteric modulation, the methodological aspect of allosteric ligand characterization, and the advantages and disadvantages of allosteric ligands compared to the orthosteric ones. This section can help the reader to get into the complex world of allosterism. The second part starts with a description of the mechanism of AR allostery and continues with the medicinal chemistry of allosteric modulators directed toward all four AR subtypes, Structure– activity relationships and the therapeutic potential of each class have been discussed, pointing out the most recent advances in the development of AR modulators. The discovery and development of AR ligands has been improved during the last decades thanks to advances in techniques and methods which belong to the great family of “Computer-Aided Drug Design.” Chapter 9, by Moro et al., deals with the application of main computational design methods in the field of ARs. Particular attention is devoted to molecular docking and molecular dynamics as the most used structure-based methods in the adenosine field, thanks to the availability of X-ray crystal structures and Cryo-EM structures of all ARs, except the A3AR. Each computational technique is critically described pointing out its merits and drawbacks. The last paragraph deals with machine learning and artificial intelligence in drug discovery, starting from a general description of the process to its application in the AR field. A receptor of the P2 receptor family is the topic of the tenth chapter, by Antonioli et al. P2 purine receptors are classified into P2X and P2Y families, each including different receptor subtypes. The P2X receptors are ligand-gated ion channel receptors and have been classified as P2X1 to P2X7. This chapter deals with the P2X4

viii

Preface

subtype, an ATP-gated cation channel that allows Na+, K+, and Ca2+ ion fluxes, and is widely expressed in cells of the immune system. The structure and molecular biology of this receptor, and its role on the functions of immune cells and microglia is reviewed. The potential of the P2X4 receptor as a drug target to treat disorders, such as neuropathic pain, neurodegenerative disease, or metabolic syndrome, is discussed. Two chapters of the book, Chaps. 11 and 12, from Supuran’s group deal with CD73 ligands in drug design campaigns for identifying antitumor agents (human enzymes) and as potential antibacterials (case in which CD73 of bacterial origin are considered). These enzymes are ectonucleases that hydrolyze ADP and less efficiently other nucleotides too, through a zinc ion-dependent mechanism. Their 3D structures are known and in the case of the mammalian enzymes this led to the design of potent inhibitors, some of which are in clinical trials as antitumor agents. The bacterial enzymes were less investigated as drug targets but might be interesting for the development of antibacterials with novel mechanisms of action. All these aspects are dealt with in the last two chapters. Overall, the present book presents the state of the art in this fascinating field of purinergic receptors. Florence, Italy July 2023

Vittoria Colotta Claudiu T. Supuran

Contents

Once Upon a Time Adenosine and Its Receptors: Historical Survey and Perspectives as Potential Targets for Therapy in Human Diseases . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Daniela Catarzi, Flavia Varano, Sara Calenda, Erica Vigiani, and Vittoria Colotta

1

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year History of Pyrazolo[4,3-e][1,2,4]Triazolo[1,5-c]Pyrimidines (PTP) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . Filippo Prencipe, Tatiana Da Ros, Eleonora Cescon, Ilenia Grieco, Margherita Persico, Giampiero Spalluto, and Stephanie Federico

47

Adenosine Receptor Ligands as Potential Therapeutic Agents for Impaired Wound Healing and Fibrosis . . . . . . . . . . . . . . . . . . . . . . . . . . Flavia Varano, Daniela Catarzi, Erica Vigiani, Sara Calenda, and Vittoria Colotta

89

Adenosine A2A Receptor Antagonists: Chemistry, SARs, and Therapeutic Potential . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 101 Andrea Spinaci, Michela Buccioni, Cui Chang, Diego Dal Ben, Beatrice Francucci, Catia Lambertucci, Rosaria Volpini, and Gabriella Marucci A2B Adenosine Receptor as a New and Attractive Target to Treat Brain Ischemia or Demyelination . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 143 Federica Cherchi, Martina Venturini, Ilaria Dettori, Felicita Pedata, Elisabetta Coppi, and Anna Maria Pugliese A3 Adenosine Receptor Ligands: From Discovery to Clinical Trials . . . . 157 Kenneth A. Jacobson, Paola Oliva, and R. Rama Suresh

ix

x

Contents

Bifunctional Tools to Study Adenosine Receptors . . . . . . . . . . . . . . . . . . 179 China Payne, Jon K. Awalt, Lauren T. May, Joel D. A. Tyndall, Manuela Jörg, and Andrea J. Vernall Allosteric Modulators of Adenosine Receptors . . . . . . . . . . . . . . . . . . . . 223 Elisabetta Barresi, Chiara Giacomelli, Claudia Martini, Federico Da Settimo, Maria Letizia Trincavelli, and Sabrina Taliani In Silico Insights Toward the Exploration of Adenosine Receptors Ligand Recognition . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 275 Davide Bassani and Stefano Moro P2X4 Receptors in Immunity and Inflammation . . . . . . . . . . . . . . . . . . . 317 Luca Antonioli, Matteo Fornai, Carolina Pellegrini, Laura Benvenuti, Clelia Di Salvo, Vanessa D’Antongiovanni, and György Haskó CD73 Inhibitors as Antitumor Agents . . . . . . . . . . . . . . . . . . . . . . . . . . 331 Clemente Capasso, Alessio Nocentini, and Claudiu T. Supuran Bacterial Ectonucleotidases: Underexplored Antibacterial Drug Targets . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 345 Alessio Nocentini, Clemente Capasso, and Claudiu T. Supuran

Top Med Chem (2023) 41: 1–46 https://doi.org/10.1007/7355_2023_158 © The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 Published online: 8 March 2023

Once Upon a Time Adenosine and Its Receptors: Historical Survey and Perspectives as Potential Targets for Therapy in Human Diseases Daniela Catarzi, Flavia Varano, Sara Calenda, Erica Vigiani, and Vittoria Colotta Contents 1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 Brief History of Adenosine and Its Receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3 The State of the Art . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1 Adenosine Production and Metabolism . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2 Distribution of Adenosine Receptors and Their Main Physiological Effects . . . . . . . . . 3.3 Signal Transduction of Adenosine Receptors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.4 Adenosine Receptor Structures . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.5 Medicinal Chemistry of Adenosine Receptor . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.6 Pathophysiological Roles of Adenosine Receptors and Emerging Therapeutic Intervention . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 Perspectives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5 Concluding Remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

2 3 8 9 9 10 11 13 22 28 31 32

Abstract Endogenous autacoid adenosine plays important physiological roles in various tissues and organs by acting through A1, A2A, A2B, and A3 receptors. Dysregulated extracellular adenosine concentrations can cause many disorders such as inflammation, neurodegeneration, pain, and cancer, thus suggesting adenosine receptors (ARs) as interesting therapeutic targets. Medicinal chemistry studies have generated a large number of AR ligands, which have been pharmacologically characterized and optimized. Improvement in AR structural biology stimulated new computer-aided drug design approaches for drug discovery. Some AR ligands have reached clinical trials, and a limited number of them are on the market. To achieve these results, great progress has been made since the first steps in the intriguing world of adenosine and its receptors. New knowledge has been achieved, many D. Catarzi (✉), F. Varano, S. Calenda, E. Vigiani, and V. Colotta Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Università degli Studi di Firenze, Sesto Fiorentino (Firenze), Italy e-mail: daniela.catarzi@unifi.it

2

D. Catarzi et al.

issues have been solved, and new important aspects, previously ignored or underestimated, have been highlighted. In particular, but not only, taking into account important factors such as binding kinetics and biased signaling of ARs can lead to a rational design of more effective and safer drugs. Multitarget-directed ligands that combine, in the same molecule, the ability to interact with AR(s) and other target(s) are getting much attention as innovative therapeutic agents for treating multifactorial disorders such as neurodegenerative diseases and cancer. Keywords Adenosine receptor ligands, Adenosine receptors, Cancer, Inflammation, Neurodegeneration, Pain

1 Introduction Our scientific journey into the magical world of adenosine and its receptors began more than 30 years ago when the knowledge on this topic was still in its infancy. One of our first readings focused on a book chapter entitled “Purine Receptors,” written by Michael William, where the beginning of a story was told [1]. Then, we let ourselves be carried away by this boundless world ready to be amazed. Once upon a time, there was an intriguing new biological “actress” named adenosine, which made its appearance on the scene as early as the late 1920s and proved to have strong effects on humans. It influenced heart rate, not increasing, but reducing it, and producing profound hypotension. An unexpected effect, if we want to see adenosine as an intriguing lady, but certainly an effect that aroused a lot of attention from insiders. However, interest in adenosine and its potential as an antihypertensive agent rapidly declined when its short half-life was evidenced. It was not until the 1950s that adenosine was again evaluated for its physiological roles. And the story goes on. Adenosine takes care of maintaining homeostasis and for this reason, it has been identified as one of the main regulators of local tissue functions and defined as a guardian angel [2]. However, our angel adenosine, though prone to its regulatory role, also knows how to arm itself to defend tissues and organs, thus becoming functional in all situations in which the organism suffers an attack or an insult. The concentration of adenosine increases under metabolically unfavorable conditions such as tissue hypoxia or stress to counteract tissue damage [3, 4]. As the guardian angel of territory that it rules and controls to great effect, adenosine has a personal army made up of four G protein-coupled adenosine receptors (ARs), termed A1, A2A, A2B, and A3, which can be considered as its warrior angels. After having played with adenosine and its receptors for a while, we leave the world of supernatural creatures and return to the scientific and rigorous sphere in which, however, adenosine continues to play a guardian angel role. In the last 50 years, a myriad of new evidence on adenosine has been discovered that allows us to draw an increasingly accurate profile. Much new information on its pathophysiological roles and effects mediated by activation of its receptors is available. Worth mentioning is the progress

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

3

that has been made in the design of new molecules capable of interacting with its receptors and producing useful effects to counteract old and new pathologies. This is an extraordinary fairy tale. However, although adenosine plays the role of homeostatic regulator, there are conditions in which excessive and chronic production of adenosine can be harmful, leading to fibrosis, inflammation, and other pathological situations [5]. It should be remembered that the results obtained to date in this field are due to the ingenuity, perspicacity, and tenacity of many researchers as well as to their ability to inspire new talents. Successes and failures have followed each other over the years, as well as moments of great ferment alternating with periods of stagnation in research. To honor the progress and successes achieved, we can trace the journey taken by those who, full of curiosity and with a pioneering spirit, sometimes overcoming the disbelief and skepticism of their colleagues, started this adventure almost 250 years ago.

2 Brief History of Adenosine and Its Receptors The history of adenosine is related to that of all other nucleosides and more directly to that of natural purine derivatives. Purine chemistry began in 1776, with Karl Scheele, a Swedish pharmacist, who isolated uric acid from a bladder stone [6]. From then on, many scientists contributed to clarifying the structure and role of uric acid, until Emil Fischer, who prepared and utilized it to synthesize all naturally occurring purines [7, 8]. Later, he solved the structure of caffeine (1,3,7trimethylxanthine) and related compounds and denoted them as purines [9]. At the same time, Albert Kossel isolated adenine and the other nucleic acid bases [10, 11]. Many efforts were made until the mid-1920s to determine the structure of nucleic acids. However, the interest was moved away from adenosine as a xenobiotic molecule with physiological actions, rather than just a component of nucleic acids. Some progress still has to be made, such as the determination of the sugar part of nucleosides and nucleotides. At the beginning of the twentieth century, the structure of AMP was proposed and D-ribose was identified as the previously unknown sugar portion [12]. ATP was isolated in 1929 independently by two research groups, but many doubts remain about the partnership of its discovery [13]. Based on growing evidence that ATP was important in cellular energy metabolism, in the early 1930s, beef heart muscle extracts containing AMP and adenosine were considered to have medicinal value. The biological effects of adenosine were described in the late 1920s when Drury and Szent-Gyorgyi described the cardiovascular effects of intravenous administration of extracts of cardiac tissues and other organs in several mammalian species [14]. They observed profound changes in cardiac rhythm and a slowing of heart rate to complete cardiac arrest. Furthermore, a potent hypotensive action was observed secondary to dilation of coronary blood flow. Intrigued, they proceeded to the isolation of the biologically active substance which they identified as an adenine derivative, i.e., adenosine-5′-monophosphate (5′-AMP). Moreover, administration

4 Fig. 1 Methylxanthine derivatives as adenosine receptor antagonists

D. Catarzi et al. O H3C O

N

CH3 N

N N CH3 Caffeine

O

O H3C O

N

NH N N CH3

Theophylline

HN O

CH3 N

N N CH3

Theobromine

of a mixture of 5′-AMP and adenosine caused qualitative and quantitative effects comparable to those produced by heart extracts. The potential of adenosine as an antihypertensive agent was attractive. IG Farben preparation, named Lacarnol, was studied, confirming that purine nucleosides and nucleotides produced profound vasodilatory effects on coronaries [15] and changes in blood pressure [16]. In these studies, it emerged that adenosine was more potent than its nucleotides in determining vasodilation or inducing hypotension. Its short half-life prevents side effects mediated by A1AR activation, such as cardiac block. The Locarnol extract was also evaluated in humans with heart disease, but the clinical trials failed and were stopped due to the lability of the natural nucleoside [17]. The study of Drury and Szent-Gyorgyi [14] can be considered as the starting point for both investigations on the physiological roles of adenosine and clarification of the biology of its receptors. However, 30 years passed before Berne and Gerlach, through independent experiments, proved that adenosine, produced by the degradation of ATP, caused hypoxic coronary vasodilation [18, 19]. However, their hypothesis has been the subject of discussion and controversy within the scientific community for several years. From then on, much research on the metabolism and physiological roles of adenosine was conducted. In the 1970s, the role of adenosine as an extracellular signaling molecule was proposed [20]. It was clarified that adenosine existed both in normally oxygenated and hypoxic myocardium [21, 22] and later its roles as a marker of hypoxia were confirmed leading to evidence of its potential effects on tissue protection [23]. At the same time, adenosine was postulated to exert its effect by its acting on specific receptors that stimulated or inhibited adenylate cyclase (AC). In particular, the induction of large accumulations of cyclic AMP in brain slices by adenosine was demonstrated [24, 25]. Moreover, the methylxanthines caffeine and theophylline (1,3-dimethylxanthine) (Fig. 1) showed to antagonize the effects of adenosine. Thus, the existence of ARs linked to cAMP production was hypothesized, and the natural methylxanthines, including theobromine, were accepted as antagonists at these receptors [4]. This study opens the door to a new vision of the nucleoside adenosine as a modulator of brain functions [24]. Other evidence had sparked interest in adenosine as a physiological regulator. One of the most incisive was the observation that the effects of exogenous adenosine can be enhanced by the adenosine uptake inhibitor, dipyridamole, which was introduced to the market in the 1950s [26–28]. In these years of great ferment, the pathways that lead to the formation and degradation of purine were also identified [29–31]. The confirmation that the enzyme responsible for the degradation of adenosine, that is, adenosine deaminase,

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

5

was localized in limited areas of the brain, strengthened the hypothesis of the role of the modulator of the nucleoside itself [32]. Until now, the physiological role of nucleosides and purine nucleotides has been increasingly emerging, often leading to heated debates. Burnstock developed his idea on the new concept of purinergic signaling that challenged the skepticism of many colleagues [20]. In 1978, he proposed the first classification of purinergic receptors based on their sensitivity to adenine nucleosides and nucleotides. In particular, the P1 receptors were the most sensitive to adenosine and AMP, modified AC activity leading to altered cAMP levels, and were competitively antagonized by methylxanthines. In contrast, P2 receptors that had high sensitivity to ATP and ADP were not antagonized by methylxanthines and caused an increase in prostaglandin production [33]. Contemporarily, progress was being made on the delineation of the adenosinergic system. In the late 1970s, the existence of specific receptors for adenosine was increasingly accepted and two AR subtypes were described, termed A1 and A2, based on their opposite effect on AC activity [34]. After the hypothesis of Sattin and Rall [24], Van Calker and co-workers confirm the existence of two different ARs [35]. In particular, the authors demonstrated the inhibitory effect of adenosine on cAMP production, while the Sattin and Rall experiments had previously shown its stimulatory action. On this basis, the receptors were classified as the inhibitory receptor A1 and the stimulatory A2 receptor. Almost simultaneously, Londos and co-workers pointed out that adenosine-like compounds exhibited different potency at the two receptors, which were classified in Ri and Ra based on the receptormediated effect on AC activity [36]. However, in the same period, effectors other than AC were identified, such as ion channels and phospholipases [37–39]. The discovery of high and low affinity receptors that activate AC led to further subdivision of the A2 subtype [40]. The high affinity receptor was termed A2AAR, and the low affinity subtype was termed A2B AR. The effects of adenosine on cAMP formation, especially in brain tissue, have been extensively studied. In the late 1970s, two groups independently demonstrated that in a specific region of the brain, rich in dopamine, AC was activated by low concentrations of adenosine analogs. This was in contrast to the results obtained in cortical brain slices where a high level of adenosine analogs was necessary to activate AC through the A2 receptor [41, 42]. Daly and co-workers confirmed the existence of the two A2A and A2B receptors based on binding studies on caffeine and related methylxanthines [43]. During the molecular cloning experiments of the adenosine A1, A2A, and A2B receptors, a new functional receptor, different from those already known, was identified and classified as the A3 receptor. The existence of A3AR was hypothesized in the mid-1980s [44] but it still took a few years before Ali and co-workers unquestionably characterized the A3 subtype as the fourth AR [45]. The receptor finally cloned [46] was different from the putative A3AR first isolated by Ribeiro [44]. In the following years, all the AR known today were cloned from several species, including humans. A report by the International Union of Basic and Clinical

6

D. Catarzi et al.

Pharmacology on the nomenclature and classification of ARs was published at the beginning of the 2000s [47] with an update in 2011 [48]. ARs belong to the family of G protein-coupled receptors. The A2A and A2B receptors were defined based on their ability to stimulate AC. Thus, they preferably interact with members of the Gs family of G proteins. As a consequence, an increase in cAMP levels was determined, leading to the activation of a pool of mediators, depending on the signaling triggered by cAMP in specific cells. In contrast, the inhibitory A1 and A3 ARs were supposed to interact with Gi/o proteins [35, 40]. However, other G protein interactions have also been described [47]. Each AR is characterized by specific secondary signal transductors which can vary in receptor tissue localization, thus influencing specific physiological and/or pathological effects. Different and new signal transduction mechanisms associated with the different ARs have been identified and clarified over the years [49]. This peculiarity opens new frontiers for future drug development. The detection and tissue localization of ARs has been facilitated and made possible by the development of specific radioligands for the different ARs. The first attempt dates back to 1978 when [3H]-adenosine was used for binding studies in fat cells [50]. The experiments failed due to concurring factors, the first among all the rapid metabolism of the nucleoside. Two years later, several radioligand binding studies on A1AR were conducted independently, leading to confirmation of the proposed classification. Labeled adenosine-like compounds and xanthine derivatives such as [3H]-CADO ([3H]-2-chloroadenosine) and [3H]-DPCPX ([3H]-8cyclopentyl-1,3-dipropylxanthine), respectively (Fig. 2), were used to bind the A1 receptor [51–53]. Radioligand binding studies on the A2A receptor were much more difficult as the first experiments failed due to high non-specific binding. The first positive results appeared in 1986 when Bruns eliminated the undesirable A1 component of a labeled non-selective A2A agonist, using an A1-selective ligand [40]. Greater success was achieved over time as new selective A2AAR agonists and antagonists, such as [3H]CGS21680 ([3H]-3-(4-(2-((6-amino-9-((2R,3R,4S,5S)-5-(ethylcarbamoyl)-3,4dihydroxytetrahydrofuran-2-yl)-9H-purin-2-yl)amino)ethyl)phenyl)propanoic acid) and [3H]-Scheme 58261 ([3H]-7-(2-phenylethyl)-5-amino-2-(2-furyl)-pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine), respectively (Fig. 2), were developed [54– 56]. One of the first radiolabeled agonists for the A3AR was [125I]-AB-MECA (Fig. 2), endowed with high specific radioactivity (>140 Ci/mmol) as a radioiodinated compound [57]. This peculiarity allowed the detection of receptors expressed in tissues at low levels. Successively, [3H]-MRE 3008-F20 ([3H]-(5-N(4-methoxyphenylcarbamoyl)amino-8-propyl-2-(2-furyl)pyrazolo[4,3-e]-1,2,4triazolo[1,5- c]pyrimidine) (Fig. 2), a high affinity radioligand antagonist for human A3AR, was reported in 2000. Both radiolabeled ligands represented a useful tool for further characterization of the A3AR subtype [58]. Until now, no selective tritiated-A2BAR agonists had been reported. [3H]-NECA 3 ([ H]-5′-N-ethylcarboxamidoadenosine) is currently the only useful radioligand to determine the affinity of ligands at this AR subtype. A tritium-labeled form of a

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . . 3

NH2 N

3

N

H

N

3

N

N

HO

O

N

N

N

N EtHN

3

O H

NH

[3H]-CADO

O OH

N

[3H]-DPCPX

3

H

H

(A1AR antagonist)

(A1AR agonist)

H

NH

N

O O

H

OH OH

H 3

H

Cl

3

NH2

O

H

7

[3H]-CGS21680 OH OH

(A2AAR agonist) 125

3

I

H

HN

3

H

NH2 N

N

N N

N

3

H

MeHN

N

N N

OH OH

(A2AAR antagonist)

H 3CO

N

O O H

[3H]-SCH58261

[125I]-AB-MECA (A3AR agonist)

NH2

O NH NH N 3

H

N N

NH2

N

O

N

3

N

H

N N

O

N EtHN

O O

N H

N

3

H

OH OH

3

H

3

[ H]-MRE 3008-F20 (A3AR antagonist)

[3H]-NECA (non-selective AR agonist)

Fig. 2 Some of the earliest AR radioligands used for in vitro studies

non-adenosine-like A2BAR partial agonist, belonging to the 3,5-dicyanopyridine series, was synthesized and evaluated as a radioligand binding probe. The study failed probably due to the high non-specific binding and moderate affinity of the ligand at the A2B receptor. However, the results obtained allowed us to hypothesize that nucleoside and non-nucleoside A2BAR agonists bind to different conformations of the receptor [59]. Many efforts have been devoted to the development of highly selective radioligands for each AR subtype, both agonists and antagonists. The most representative examples have recently been reported in some reviews which are recommended for further information on chemical probes for ARs [60, 61]. The availability of highly selective AR radioligands helped not only the tissue localization of the different ARs but also the clarification of their functional coupling and regulation. In addition to radioligands useful for the in vitro characterization of receptors, also specific chemical probes for ARs were developed, which are useful for drug discovery, diagnostics, and for application to structural biology studies [60]. Currently, the application of the methodology for covalent probes represents an important tool for stabilizing target proteins to obtain X-ray crystal structure [48, 61, 62]. All of these advances have been made possible by the intense development of a large number of AR agonists and antagonists, both by pharmaceutical industries and

8

D. Catarzi et al.

academic research groups. Although the first studies date back to the 1950s [28], the following years represented a period of great ferment for the identification of new pharmacological tools. Among the first compounds studied, the adenosine-like derivatives emerge mainly as agonists and the xanthine derivatives are among antagonists [63]. Many non-xanthine compounds have also been designed to antagonize the ARs. Bicyclic antagonists with a purine core were developed by replacing the ribose of adenosine with smaller substituents. In fact, in the late 1980s, it was evidenced that the elimination of the ribose moiety from the adenosine structure led to AR antagonists [64]. This experimental observation consolidated the idea that the presence of the ribose moiety was essential for agonist activity. William and co-workers reported a series of new 1,2,4-triazolo[1,5-c]quinazolines that paved the way for many derivatives with a tricyclic structure [65]. Since then, hundreds of new AR ligands endowed with different degrees of affinity and selectivity have been synthesized and extensively reviewed [4, 63, 66– 77]. More recent achievements are related to the clinical development of AR ligands and also their marketing [77, 78]. A great deal of progress has been made since 1985, when adenosine was approved as a drug to treat supraventricular tachycardia (Adenocard®) [79] and, later, to act as a vasodilatory agent in myocardial perfusion imaging (Adenoscan®) [80].

3 The State of the Art The historical survey carried out to synthesize the most important milestones highlights how difficult it was to reach the current level of knowledge on adenosine and its receptors. To date, we know that adenosine is a ubiquitous extracellular signaling molecule that produces its effects through the activation of G proteincoupled ARs that are divided into four subtypes, termed A1, A2A, A2B, and A3 [49]. The widespread distribution of its receptors indicates that adenosine plays a fundamental role in the regulation of many important functions in tissues and organs. Under physiological conditions, extracellular adenosine reaches a concentration of 30 to 200 nM, which is high enough to activate both the A1 and A2A ARs. On the contrary, under stress conditions such as hypoxia or high metabolism, the level of extracellular adenosine can grow to very high micromolar values, thus activating also low affinity A2B and A3 subtypes. Thus, adenosine can be considered as a homeostatic regulator or, more correctly, a “retaliatory metabolite,” the role it has earned in the past [81].

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

3.1

9

Adenosine Production and Metabolism

At the extracellular level, adenosine is generated from nucleotide (i.e., ATP, ADP, and AMP) dephosphorylation, mainly through the activation of ectonucleotidases CD39 and CD73 [49, 82]. Adenosine precursors are released by different cells under stressful conditions and then transformed into the protective mediator adenosine. Under physiological conditions, adenosine can also be generated intracellularly or transported within the cell from the extracellular space. In particular, hydrolysis of S-adenosylhomocysteine or AMP through cytosolic S-adenosyl-homocysteine hydrolase and endo-5′-nucleotidase, respectively, leads to the formation of adenosine [83]. Otherwise, because of different types of nucleoside transporters, adenosine can cross the cell membrane with different mechanisms and increase its intracellular concentration. Obviously, in stressful conditions such as hypoxia, the direction of adenosine flux can be reversed [84]. In extracellular spaces, adenosine is transformed into inosine by adenosine deaminase (ADA) or phosphorylated to AMP by adenosine kinase (AK). Adenosine that has not been taken up or deaminated interacts with its ARs.

3.2

Distribution of Adenosine Receptors and Their Main Physiological Effects

The wide distribution of ARs throughout the body highlights the important physiological roles, both in the central nervous system (CNS) and in the periphery, attributed to adenosine [85]. Each receptor is characterized by a peculiar distribution at the tissue and organ level and, sometimes, by multiple secondary signal transductors that can mediate a plethora of physiological effects. Additionally, ARs show different degrees of affinity for adenosine, ranging from the highest, typical of the A1AR (Ki = 70 nM), passing through the intermediate of the A2A receptor (150 nM) to the lowest of the A2B and A3 ARs (5,100 and 6,500 nM, respectively). The affinity of adenosine for its receptors is modulated by ectoadenosine deaminase (ecto-ADA). In particular, ecto-ADA interacts with specific binding sites on the ARs leading to increased receptor affinity and signaling [86, 87]. The A1AR is highly represented in different areas of the CNS (brain cortex, hippocampus, cerebellum, spinal cord, and glial cells) [83], a distribution that justifies the wide range of physiological effects mediated by it such as anticonvulsant, anxiolytic, pain reduction, sleep/wakefulness cycle control, modulation of neurotransmitter release [88, 89]. This subtype is also widely distributed in the cardiovascular system with the major expression in the heart, where it exerts negative inotropic, chronotropic, and dromotropic effects. In adipose tissue and pancreas, A1AR modulates glucose and lipid metabolism by inducing inhibition of lipolysis and secretion of insulin, respectively [90]. At the kidney level, where it is located in cells of the juxtaglomerular apparatus, collecting ducts of the papilla and

10

D. Catarzi et al.

inner medulla, the A1 subtype reduces renal blood flow, glomerular filtration rate, and renin release [91, 92]. It is also distributed in the respiratory system (airway epithelium and smooth muscles), where it produces bronchoconstriction, and in the immune system (neutrophils, eosinophils, macrophages, and monocytes) where its main effects are pro-inflammatory [93, 94]. Furthermore, the A2AAR subtype is widely distributed in the body, but with the highest expression in the central nervous (striatum, olfactory tubercle) and immune systems. Lower levels of this subtype were found in other CNS districts (cerebral cortex, hippocampus) and the heart. In the peripheral immune system, the A2AAR, located in leukocytes, mediates numerous anti-inflammatory effects. This receptor subtype was identified as an important mediator of adenosine inhibition of platelet aggregation [95]. A2BAR is less expressed in the brain than in the periphery, where it is highly expressed in the bowel, bladder, lung, and also in different types of cells such as fibroblasts, platelets, endothelial and immune cells. This subtype is gaining a prominent role as a modulator of inflammation and immune responses in many diseases including cancer, diabetes, and other vascular disorders. Moreover, its upregulated expression in selected stressful conditions (hypoxia, inflammation) underlines its pathophysiological relevance [5]. The A3AR subtype has a low concentration in the brain, where it can be found in restricted areas, including the cortex, hippocampus, thalamus, and hypothalamus, as well as motor nerve terminals. It is also expressed in selected cells and tissues such as microglia and astrocytes, through which it mediates the neuroinflammatory response directly associated with a reduction of neuropathic pain [96]. Controlling of inflammation was also evidenced at the peripheral level where the A3 receptor is located in inflammatory cells such as lymphocytes, eosinophils, neutrophils, monocytes, macrophages, and mast cells, which are involved in the A3AR anti-inflammatory effects [97]. Overexpression of A3AR in different cancer cells and tissues is also reported, thus attributing an important antitumoral role to this receptor subtype [98]. At the periphery, A3AR is also distributed in other districts, including enteric neurons, lung parenchyma, bronchi, and, not less important in the heart (coronary and carotid arteries) where it mediates cardioprotective effects [99].

3.3

Signal Transduction of Adenosine Receptors

Specific signal transduction systems and a great variety of switchable-associated mediators are connected with each AR, which mediates physiological responses also depending on the type of cell or tissue involved. The A1 and A3 ARs are coupled to heterotrimeric G proteins belonging to the Gi/Go family, while the A2A and A2B receptors predominantly signal through Gs proteins. Gi/Go proteins inhibit AC, thereby lowering cAMP levels (A1 and A3 ARs), and increase both inositol

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

11

1,4,5-trisphosphate (IP3) and intracellular Ca2+ levels by activating phospholipase C (PLC) via Gβγ subunits (A3AR). In contrast, activation of Gs proteins leads to a cAMP level increase by stimulation of AC activity. Nevertheless, increasing evidence suggests that ARs can couple to multiple G proteins [49, 100, 101]. For example, A3 and A2B receptors are also coupled to Gq proteins, whose stimulation activates PLC and produces an increase in calcium levels [48, 49]. A2AAR is also reported to couple to AC via Golf in the striatum [102]. All ARs are involved in the intracellular phosphorylation cascade of the mitogen-activated protein kinase (MAPK) family, giving them a role in cell growth, survival, death, and differentiation [49, 103]. More recently, it has been evidenced that ARs can also signal through G proteinindependent effectors [49]. In particular, one of these G protein-independent pathways, with β-arrestins as signal transducers, was demonstrated for the A2B and A3 ARs, with limited evidence for the A1 and A2A subtypes [104]. The identification of G protein-independent pathways other than those coupled to G proteins and associated with a specific AR, allowed to open a new frontier for the development of drugs with fewer side effects. In fact, each signal pathway, associated with a specific receptor subtype, can produce different effects depending on the cell type or tissue. Therefore, the availability of AR agonists, defined as biased, able to distinguish between the different signaling pathways and to choose one over the others, could allow obtaining targeted therapeutic actions. In particular, avoiding pathways associated with undesirable effects could lead to increased drug safety. A first step forward seems to have been made. Wall and co-workers highlighted the importance of the concept of biased agonism. An A1AR-selective agonist, which was reported to activate specific G proteins, was demonstrated to be a potent analgesic devoid of side effects at cardiovascular, respiratory, and CNS levels [105].

3.4

Adenosine Receptor Structures

The four receptor subtypes were purified and successfully cloned from mammalian and non-mammalian species, particularly rats, mice, and humans [47]. Considerable species differences were observed in some cases, which are more pronounced for the A3 subtype [106, 107]. Based on sequence similarity and G protein-coupling specificity, A1 and A3 receptors share 49% sequence while A2A and A2B receptors are almost 59% identical. The most conserved region may be found in the extracellular loops of the receptor where the homology sequence reaches 71%. All ARs are characterized by a common structure consisting of seven transmembrane α-helices (7TM, helices 1–7), an extracellular amino-terminus (N-terminus), a cytosolic carboxy-terminus (C-terminus), three extracellular loops (EL1-3), and three intracellular loops (IL1-3) of different lengths and functions, depending on the AR subtype [47, 108]. These domains confer specific characteristics that are important for receptor–ligand interactions. The N-terminus has one or more glycosylation sites, and the C-terminus contains phosphorylation and palmitoylation

12

D. Catarzi et al.

regions, which are involved in both receptor desensitization and internalization processes. The complex structures of A1 and A2A ARs were determined using X-ray crystallography, NMR techniques, or, more recently, cryo-electron microscopy [78]. The first investigations that led to the elucidation of the A2AAR structure through X-ray crystallography [109] helped the other AR architecture to be solved. Other studies include crystal structure determinations of complexes with antagonists [109, 110] and agonists [111, 112], and for a ternary complex of A2AAR with an agonist and a mimetic engineered G protein [113]. Regarding A1AR, many molecular modeling and mutagenesis studies have been conducted before crystal structures of the A1AR were proposed [114, 115]. The first simulations were performed computationally using homology models of the rhodopsin or A2AAR structure [116]. In 2017 Glukhova et al. proposed a crystal structure of the A1AR bound to a covalent xanthine antagonist [117]. The computational and mutagenesis analysis of the A1AR identified peculiar differences from the previously solved A2AAR structure. In particular, a distinct conformation of the second extracellular loop and a wider extracellular cavity endowed with a secondary binding pocket for orthosteric and allosteric ligands were highlighted. Crystallization of a receptor, often necessary for structural biology studies, is still a very challenging task. In fact, referring to ARs, their low expression in native tissue, and their instability once extracted from the membrane, can represent important limitations. More recently, cryogenic electron microscopy (cryo-EM) has attracted wide attention as an innovative approach for receptor structure elucidation without the use of crystallization. By using this innovative technique, many detailed structures of A2AAR have been reported over the years. In fact, the A2A has become one of the most investigated GPCR structures [118, 119]. In addition, structures of the A1 and A2A ARs, coupled with the suitable G protein or with its engineered parent, have been determined. These are the only examples of GPCR-G protein complex structures reported in the literature till now. In particular, the adenosine-bound A1AR coupled with the Gi protein [120] and the NECA-bound A2AAR coupled with an engineered Gs protein [121] were studied. The complex structures provided important insights into the active conformations of the A1 and A2A ARs and the G proteins coupled to them. However, the dynamic mechanism of the specific interactions remains unclear. Studies performed to clarify the AR structure using advanced techniques (X-ray crystallography, cryo-EM, and NMR) have generated several structures for ARs. Their availability has stimulated different new structure-based and computer-aided drug design (CADD) approaches for the development of new AR ligands [122]. To date, the Protein Data Bank (PDB) [123] has recorded approximately 64 structures of ARs under different functional states. However, structural studies on A2B and A3 AR have not yet led to the desired results. Furthermore, there is a major limitation regarding the structural studies of all AR subtypes. In fact, there are no experimental techniques available that can evaluate the

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

13

flexibility of the receptors, which plays a critical role in the regulation of their biological functions. Moreover, only static images are available in PDB structures that cannot capture and describe the dynamics of the ligand–receptor interaction. However, molecular dynamics (MD) simulations [124] seem to be useful for this purpose, providing models of the ligand-binding/dissociation processes and new insights into the functional mechanisms of ARs. The Gaussian accelerated molecular dynamics (GaMD) method has been used to clarify the mechanism of A1 and A2A AR–G protein interactions [125]. A review of simulation studies has recently been reported which emphasizes the high potential of these types of techniques for the rational design of drugs targeting ARs [126].

3.5 3.5.1

Medicinal Chemistry of Adenosine Receptor Adenosine Receptor Ligands, in Short

ARs are widely distributed throughout the body and their activation modulates a wide variety of effects in different tissues and organs. Thus, the regulation of their activity could have therapeutic potential, but at the same time, open the possibility of side effects occurring. For these reasons, the development of selective agonists and antagonists is particularly intriguing. However, modulation of AR activity can be realized not only by using orthosteric ligands but also by allosteric modulators. Allosteric ligands recognize a distinct binding site, conformationally linked to that of orthosteric ligands [127]. The therapeutic utility of AR allosteric modulators, in particular positive allosteric modulators (PAMs), has been established in various preclinical animal models. In this regard, a reading of recently published in-depth reviews on AR allosteric enhancers also focused on selected ARs [4, 68, 128, 129] is recommended. Over the years, different classes of ligands have been evaluated at ARs because of the availability of potent and selective radioligands which emerged as essential to support the drug discovery process. In fact, the first screening of newly synthesized compounds consists of a rapid radioligand binding assay. Together with functional assays, useful for evaluating the pharmacological profile of ligands, binding studies have helped define the recurring structural features that can affect the affinity or efficacy of compounds for a specific AR [4]. This was not an easy challenge in the case of AR antagonists, as a result of both the large number and variety of their scaffolds that made it difficult to identify common structural requirements. Although antagonists are characterized by large structural variability, the agonist profile has been associated for a long time with an adenosine-like structure.

14

3.5.2

D. Catarzi et al.

Emerging Adenosine Receptor Ligands as Tool Compounds

When the biological relevance of adenosine emerged, the availability of specific tool compounds for AR studies was an important goal to be achieved. Subsequently, the discovery of specific AR radioligands, and also fluorescent and covalent ligands, all useful to facilitate receptor characterization, structural biology studies, drug discovery and diagnostics, emerged [60, 61]. The AR agonists and antagonists collected in the following can be considered as some of the main players that have contributed greatly to tool compounds to deepen the knowledge of the world of adenosine and its receptors. AR affinities of the reported compounds can be found in the references cited.

Agonists Adenosine itself has been used for therapeutic and imaging purposes for several years. Although the rapid metabolism of adenosine in the bloodstream has allowed its use as a diagnostic tool or agent in emergency therapy, nevertheless it has also represented a disadvantage for other potential applications. In the past decades, the medicinal chemistry approach to obtain AR agonists was to perform selected structural modification at N6- and 2-positions of the adenosine nucleus, and/or at the 3′-, 4′- or 5′-positions of the ribose moiety. Efforts to synthesize more stable analogs began in the 1960s, producing some compounds including 2-chloroadenosine (CADO) and NECA [130] (Fig. 3). Suitable substitutions of the adenosine core led to AR agonists with varying degrees of subtype selectivity. Much effort has been made to obtain clear and complete structure-activity relationships (SARs) of adenosine-like derivatives [4, 63, 66–68, 70–74] which have helped the discovery of more suitable compounds to define the pharmacology of AR subtypes. Although the adenine nucleus can be decorated with more flexibility to yield potent and selective AR agonists, modifications at the ribose moiety require particular attention. In fact, structural alterations at this level can produce a modulation not only of AR affinity and selectivity, but also of the efficacy of the target compound. The rationale for these modifications was first to obtain compounds endowed with subtype selectivity by changing the electronic and steric features of the molecules. A challenge was to improve the physicochemical properties of compounds, with the improper pharmacokinetic profile often being a drawback in promoting a drug candidate to a therapeutic agent. Some AR agonists were obtained through substitution at the adenosine N6-position. An example is the N6-cyclopentyl substituted derivative CPA (N6-cyclopentyladenosine) which was shown to selectively bind to the human A1AR. CADO is a metabolically stable analog of adenosine that behaves as a non-selective AR agonist. It has been used to define the AR pharmacological profile in a particular cell/tissue. Tritiated CADO was used as a radioligand probe for AR studies [131].

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

NH2

NH2 N

N

N

N

N

HO

EtHN

H

H

CADO (A1AR)

OH OH

O O

MeHN H

IB-MECA

OH OH

(A3AR)

NH2 N

N

N

N

N

O O H

N

O O

I

N MeHN

N

N

HN

N

Cl

N N

N

HO O H

Cl-IB-MECA

OH OH

OH OH

(A2AAR)

NH2

NH2 CN

N S CN

CN

N NH

O

S

N

O

CN

OCH 3

LUF 5845

BAY 60-6583

(non-selective)

(A2BAR)

NH2 N

N

CN

N

Cl

N CN

O

CN

N

Cl

S

S

CONHCH 3

Regadenoson

(A3AR)

H2N

(A1AR)

I

N

CGS21680 (A2AAR)

CCPA

OH OH

(A1AR)

HN

NH

OH OH

H

CPA

OH OH

OH

N

S

OH S

CN

Capadenoson

Neladenoson

(A1AR)

(A1AR)

O

NH2 N

Cl

N S

N

N

CN

N

Cl

O

O

N

N

H

H

NECA

N

HO

O

OH OH

N

N

N

HO

(non-selective)

N

N

N

N

NH2

EtHN

N

N

O O

O

HN

HN

N

N

Cl

15

N HO

O

S CN Neladenoson bialanate

O

O

NH O

(A1AR)

Fig. 3 Adenosine receptor agonists as tool compounds

N

OCH 3

O NH2

H

OH OH

Spongosine (A2AAR)

OH

16

D. Catarzi et al.

2-Chloro-N6-cyclopentyladenosine (CCPA) displayed slightly higher human A1AR affinity than the parent compound CPA. Among the first adenosine-like compounds that appeared, the 5′-N-ethylcarboxamido-derivative NECA behaved as a broad-spectrum AR agonist, with low affinity for the A2B subtype [40]. However, it has been one of the most widely used AR agonists, especially as a useful tool for studying the A2B subtype, which lacks potent and selective agonists with high efficacy [132]. Substitution with the small 5′-N-alkyluronamide group on the ribose moiety of adenosine as in NECA, in general, provided increased potency at all ARs. This structural feature is present in the first selective A2A receptor agonist CGS21680 (2-[4-(2-carboxyethyl)phenylethylamino]-5′-Nethylcarboxamidoadenosine), which was shown to be potent at A2AAR in rat and mouse, but with lower selectivity in humans. This 2-(2-phenylethyl)amino modification was particularly effective in improving affinity at the A2A subtype. CGS21680 was tested in a Phase 2A clinical trial as a potential antihypertensive agent. However, administration caused a large fall in blood pressure and consequent increase in cardiac output. The introduction of a 3-iodophenyl moiety at the N6 position in the NECA structure led to the A3AR agonist IB-MECA (Piclodenoson, 1-deoxy-1[6-[[(3-iodophenyl)methyl]amino]-9H-purin-9-yl]-N-methyl-β-Dribofuranuronamide), whose selectivity versus the A1 and A2A subtypes was very low. Its 2-chloro analog Cl-IB-MECA (namodenoson, 1-[2-chloro-6[[(3-iodophenyl)methyl]amino]-9H-purin-9-yl]-1-deoxy-N-methyl-β-Dribofuranuronamide) emerged as a very potent and highly selective A3AR agonist. Both IB-MECA and its parent compound Cl-IB-MECA have been in clinical development. Regadenoson (CVT3146, Lexiscan®)(1-[6-amino-9-[(2R,3R,4S,5R)3,4-dihydroxy-5-(hydroxymethyl)oxolan-2-yl]purin-2-yl]-N-methylpyrazole-4carboxamide), though not a highly selective A2AAR agonist in humans, entered clinical trials for many diseases (transplantation and lung inflammation). Many other new and old AR agonists have been and are being clinically evaluated for human use [77, 78]. In fact, from the first results, over the years many AR agonists, endowed with high affinity and selectivity, have been reported that were modified differently at the ribose moiety and substituted at the N6 and C2 adenine positions [4, 63, 66– 68, 70–74, 77]. The positive results obtained are an expression of all the efforts made by the scientific community to obtain drug candidates with high efficacy and a suitable pharmacokinetic profile. Although the agonist profile has long been associated with an adenosine-like structure, more recent studies reported some non-nucleoside compounds belonging to the 6-aminopyridine-3,5-dicarbonitrile series that showed different degrees of efficacies at the different ARs (Fig. 3). Some of these ligands, although lacking the ribose portion, have significant affinity and efficacy for human ARs. For example, BAY60-6583 (2-[[6-amino-3,5dicyano-4-[4-(cyclopropylmethoxy)phenyl]-2-pyridinyl]thio]-acetamide) [75] and LUF 5845 (2-amino-4-(4-methoxy-phenyl)-6-(1H-imidazol-2-ylmethylsulfanyl)pyridine-3,5-dicarbonitrile) [75, 133] are among the prototypical 6-aminopyridine3,5-dicarbonitriles reported in the literature.

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

17

Although BAY60-6583 is a potent and selective human A2BAR agonist discovered by Bayer Healthcare, LUF 5845 and its analogs displayed in general nanomolar affinity for all ARs, and in some cases even considerable efficacy. BAY 60-6583 behaves as an A2B partial agonist [134], but showed to act as an antagonist at the other ARs [106]. In fact, the pharmacological profile of this compound can change depending on the tissue, receptor expression level, and in situ adenosine concentration. Amino3,5-dicyanopyridine compounds have common features, that is two cyano groups at C3 and C5 and a thiomethyl group with an imidazole-2-yl moiety at the 6-position. The 4-phenyl moiety can be substituted with a suitable group (H, OH, OCH3, OCH2cC3H5) in the para- or meta-position. Modification of LUF series compounds at the level of thiomethyl-side-chain moiety led to the development of a potent and selective partial A1AR agonist which was developed as a radioligand for positron emission tomography (PET) imaging in the brain [77, 135]. BAY 60-6583 was introduced as a selective non-nucleoside A2BAR agonist with high affinity/potency, depending on the type and condition of the assay [134, 136, 137]. It was used for several pharmacological studies, thus allowing its beneficial effects in obesity [138] and insulin resistance to emerge [139, 140], but also cardiovascular disorders [141, 142]. The exhaustive review of Dal Ben and colleagues [75] describes developments in this field from 2000 to recent times. The lucky structural modifications of dicyanopyridine derivatives led to the development of compounds with a substituted thiazole group linked to the scaffold at the 2/6 position through a thiomethyl function. Capadenoson (2-amino-6[[2-(4-chlorophenyl)-1,3-thiazol-4-yl]methylsulfanyl]-4-[4-(2-hydroxyethoxy)phenyl]pyridine-3,5-dicarbonitrile) is currently the most representative compound of this series, as it represents a reference A1AR partial agonist, endowed with sub-nanomolar activity at the human A1AR [143]. Capadenoson has been shown to be useful in different stressful and diseased conditions [144–148] showing a favorable pharmacokinetic profile [146]. Moreover, the advantages of the use of a partial A1AR agonist are primarily based on the lower risk of producing severe side effects compared to full agonists [146, 149]. Structural modifications at the exocyclic amine group of Capadenoson led to the development of other dicyanopyridine derivatives whose lead compound was the A1AR agonist Neladenoson (2-(((2-(4-chlorophenyl)-1,3-thiazol-4-ylmethyl)sulfanyl)-4-(4-(2-hydroxyethoxy) phenyl)-6-(pyrrolidin-l-yl)pyridine-3,5-dicarbonitrile). It is a selective A1AR partial agonist [150, 151] with a pharmacological profile similar to that of Capadenoson, but endowed with lower central undesirable effects. Its prodrug, in the form of its bialanate derivative (alanine–alanine ester), Neladenoson bialanate (BAY 1067197), was developed that led to an improvement in the pharmacokinetic profile. Analogs of Capadenoson were designed and synthesized, focusing attention on their dissociation kinetic properties at the A1AR [152]. Some of these derivatives showed a residence time longer than that of Capadenoson. Recently, a thorough review of current and historical AR agonists in preclinical and clinical development has appeared [77].

18

D. Catarzi et al.

Antagonists In general, antagonists are nitrogen-containing heterocycles endowed with a planar π-electron rich structure, generally decorated with hydrophobic groups that modulate not only AR affinity but even selectivity. Finally, they lack ribose moiety (Fig. 4). The prototypical AR antagonists are caffeine and theophylline, natural psychostimulant xanthines, endowed with micromolar affinity and low selectivity for ARs (Fig. 1) [153]. Taking these latter as lead compounds, many other differently substituted xanthine derivatives were designed and synthesized yielding the definition of detailed SARs which have been confirmed over the following years [4, 154]. The introduction of suitable substituents at the 1-, 3-, and 8-positions of the xanthine scaffold shifts the affinity toward a particular receptor subtype. A very large number of selective AR antagonists have been developed as derived from natural xanthines. A1AR selective compounds typically feature a bulky cycloalkyl substituent at 8-position such as 1,3-dipropyl-8-cyclopentylxanthine (DPCPX) [18] which is, however, only moderately selective for this subtype. Alkylation of the nitrogen atom at N1 and N3 positions with N-propyl groups increases the affinity at all AR subtypes, even if it was demonstrated later that A2BAR prefers N3-unsubstituted compounds. The presence of a styryl moiety at the 8-position of the xanthine scaffold improves the selectivity for the A2A subtype as demonstrated by (E)-8(3,4-dimethoxystyryl)-1,3-diethyl-7-methylxanthine (Istradefylline, KW-6002) which is the first A2AAR antagonist approved as a drug [155]. It is highly selective versus A3 and A2B ARs but not the A1 subtype. An associated problem is its E/Z isomerization and also light-induced dimerization [156]. During the past decade, potent and selective A2BAR antagonists belonging to the xanthine series, such as PSB-603 (8-(4-(4-(4-chlorophenyl)piperazine-1-sulfonyl) phenyl)-1-propylxanthine), have been developed, that displays a Ki value of 0.553 nM at the human A2B receptor and >15,000-fold selectivity over other ARs. A tritiated form of the compound was prepared as a new radioligand and characterized in kinetic, saturation, and competition studies [157]. Further modifications at position-8 have resulted in many other highly selective AR antagonists of interest. MRS 1754 (8-[4-[((4-cyanophenyl)carbamoylmethyl) oxy]phenyl]-1,3-di(n-propyl)xanthine) is a selective antagonist for A2BAR with a very low affinity for the A1 and A3 receptors of both humans and rats, which has been prepared as a radioligand [132, 158]. More recently, many xanthine-based AR antagonists emerged, showing also promising efficacy in vitro/in vivo animal models of Parkinson’s disease (PD) [159], diuresis [160], and asthma [161, 162]. In addition, starting from the adenine structure, various non-xanthine cores were taken into consideration to develop new AR antagonists. In fact, the decoration of bicyclic and tricyclic ring systems led to the identification of highly potent and selective AR antagonists with improved affinity and selectivity with respect to xanthine derivatives (Fig. 4). 9-Chloro-2-(furan-2-yl)-[1,2,4]triazolo[1,5-c]quinazolin-5-amine (CGS15943) was one of the most potent tricyclic non-xanthine AR antagonists reported in the literature as non-selective AR ligand [65]. Despite this, it has been considered for a

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . . O N

N O

O

N

N

O O S N

NH

N

OCH 3

NH

N

DPCPX

Istradefylline

PSB-603

(A1AR)

(A2AAR)

(A2BAR)

O O

N

O

Cl O

O

N N

N

N

N

H 3CO

NH2 NH

N O

O

OCH 3

CH3 N

N N

O

O

NH

19

NH

NH

N NH

CN

N N

N Cl

MRS 1754

N N

CGS15943 (non-selective)

(A2AAR)

O

N MRE 3008-F20 (A3AR)

OCH 3

NH2 H 3CO

O

N N

N N

N

N

O

NH N

(A2AAR)

(A2AAR)

NH2 N N

O

N

N

O

O

N N N

H3C

N

N

Tozadenant

O

O

H2N

CH3

O

N

Preladenant

N

OH

S

N

N N

N

O

S

O

N

O NH2 Vipadenant

Ciforadenant

CH3

(A2AAR)

O N

N

N

(A3AR)

NH2

F

S O

MRS 1523

(A2AAR)

N

N

N N

N O NH2

Inupadenant (A2AAR)

HO

N N Etrumadenant (A2A/A2B ARs)

O O

NH2 Br

N N N

N

CH3

N

CH3 N

H3C

S

N

CN

F

N

NH

N N

O

N

N

O NH

Taminadenant (A2AAR)

FO 2S

DU172 (A1AR)

Fig. 4 Adenosine receptor antagonists as tool compounds

long time as the lead compound for the new generation of tricyclic AR antagonists of which the most representative are reported in Refs. [4, 66–69, 71–73, 76]. CGS15943 is substituted at position 2 with a furan-2-yl group, which is

20

D. Catarzi et al.

considered an important feature for A2A activity and selectivity [163]. Successively, its structural modification produced many other tricyclic AR antagonists with different degrees of affinity and selectivity at all four ARs [4, 66–69, 71–73, 76]. One of these emerging compounds is MRE 3008-F20 which is derived from the structure-activity optimization of pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine derivatives first reported by Gatta et al. [164]. It is a highly selective hA3AR antagonist with comparable activity in the cAMP functional assay and good selectivity versus the other AR subtypes [165]. The same drug design approach led to the identification of Preladenant (Scheme 420814, 2-(2-furanyl)-7[2-[4-[4-(2-methoxyethoxy)phenyl]-1-piperazinyl]ethyl]7H-pyrazolo[4,3-e][1,2,4] triazolo[1,5-c]pyrimidine-5-amine), which is one of the most potent and selective A2A receptor antagonists evaluated in a clinical trial for PD. Though it is well tolerated no significant beneficial effects were obtained following its therapeutic use. The discovery of A2AAR antagonists with acceptable selectivity, physicochemical, and pharmacological properties has continued to be an intriguing challenge [166]. Hoffmann-La Roche identified a benzothiazole-based potent and selective A2AAR antagonist, Tozadenant (4-hydroxy-N-(4-methoxy-7-morpholinobenzo[d] thiazol-2-yl)-4-methylpiperidine-1-carboxamide), which reached phase 3 clinical trials for idiopathic PD but the process was interrupted based on an unexpected emerging safety. Earlier SAR studies on bicyclic core compounds led to highly potent hA2AAR antagonists of the next generation to CGS15943. Some pyrazolo-, -pyrrolo-, and imidazo-pyrimidine derivatives endowed with high A2A receptor affinities and selectivity were obtained as Vipadenant precursors [167]. Some selected imidazo-pyrimidines showed to be orally active in a haloperidol-induced hypolocomotion mouse model assay. In addition, low oral bioavailability and poor in vivo stability were observed in rat models. By replacing the imidazole nucleus with a triazole, a series of 7-aryl triazolo[4,5-d]pyrimidine compounds was obtained. Optimization of the latter yielded Vipadenant (3-[(4-amino-3-methylphenyl) methyl]-7-(furan-2-yl)triazolo[4,5-d]pyrimidin-5-amine, BIIB014, V2006) which showed good human A2AR affinity and selectivity versus the other AR subtypes, and also improved pharmacokinetic properties. Vipadenant was demonstrated to reverse haloperidol-induced hypolocomotion in mice and rats. However, its clinical trials for PD were terminated in 2010 due to safety concerns emerging in preclinical toxicology testing [168]. Investigation of benzothiazole derivatives as Tozadenant analogs led to the discovery of a new potent and selective A2AAR antagonist endowed with high aqueous medium solubility and stability versus microsomal enzymes. Selected compounds have been shown to be effective in an in vitro PD model [169]. To address the chemical structural liabilities that probably have led to concerns about Vipadenant toxicity, further modification led to the lucky discovery by Vernalis of Ciforadenant (7-(5-methylfuran-2-yl)-3-[[6-[[(3S)-oxolan-3-yl]oxymethyl]pyridin2-yl]methyl]triazolo[4,5-d]pyrimidin-5-amine, CPI-444, V81444), which is currently under clinical evaluation for cancer therapy [78]. Further structural modification of Tozadenant yielded other A2AAR antagonists showing high affinity and selectivity for this subtype. The radiolabeling of some

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

21

fluorinated compounds with 18F and in vitro autoradiography revealed that these compounds represent potential candidates for the visualization of the A2A receptor [170]. In addition to the bicyclic scaffold, various monocyclic compounds were also reported including suitably substituted pyridines, pyrimidines, and thiazoles [4, 66– 69, 71–73, 76] (Fig. 4). For example, MRS 1523 (6-ethyl-5-[(ethylthio)carbonyl]-2phenyl-4-propyl-3-pyridinecarboxylic acid propyl ester) is an A3 AR antagonist at both human and rat. The compound is only moderately potent but very selective in humans, and, more importantly, it was the first derivative to possess sub-micromolar affinity at the rat A3ARsubtype [171]. Furthermore, MRS 1523 was useful in clarifying the involvement of the A3AR in the inhibition of melanoma cell growth. In fact, it has been shown to counteract, both in vitro and in vivo, the modulation in the expression of key signaling proteins, demonstrating that the A3AR response leads to inhibition of tumor growth [172]. MRS 1523 can exert an antihyperalgesic effect through the blockage of the N-type Ca2+ channel in isolated neurons of the rat dorsal root ganglion (DRG). This effect is in common with its ancestors, 1,4-dihydropyridines, known as calcium channel inhibitors, from which it derives by oxidation [173]. Saini et al. reported an in-depth review of AR antagonists covering literature data from 2013 to 2021 [76]. Jacobson et al. proposed a historical overview of the chemical scaffolds typical of AR ligands, highlighting those compounds, respectively, derived from xanthine, adenine, and prototypical monocyclic scaffolds [174]. The virtual screening of chemical libraries using computational approaches has contributed to the discovery of novel AR antagonists. Molecular docking studies at the four ARs, improved by the availability of the X-ray crystal structure of the A1 and A2A subtypes, have also allowed rationalizing the SARs of a large number of chemical classes of AR antagonists and highlight similarities and differences between the AR binding sites [117, 175–181]. Many compounds that act at the ARs were evaluated that showed their potential as therapeutics. Thus, numerous clinical trials have been conducted, leading to the identification of many agents capable of modulating adenosinergic signaling. Although most of these trials have not been pursued, in some cases the preliminary results were encouraging [77, 78, 182–189]. Currently, in addition to adenosine, only Regadenoson [190] (Fig. 3) and Istradefylline [156] (Fig. 4), agonist and antagonist, respectively, of the A2AAR, are approved for use in humans. In parallel to preclinical and clinical trials, many studies are in progress to investigate new promising compounds as modulators of the adenosine system. The dichotomy of the adenosine system and the consequent controversies that still exist about the pathophysiological roles of ARs make the goal more challenging and stimulating [2, 5].

22

3.6

D. Catarzi et al.

Pathophysiological Roles of Adenosine Receptors and Emerging Therapeutic Intervention

Generally, both acute and physiological increases in adenosine production are associated with beneficial effects such as vasodilation and decreased inflammation [2]. However, in important pathological states, adenosine levels can increase due to chronic overproduction, leading to harmful effects such as inflammation and organ damage [5]. Under these conditions, adenosine takes off its guardian angel clothes and becomes a bad angel. Due to the dichotomy of adenosine behavior, the adenosine system can be considered in all respects as an important and strategic target for counteracting old and new unmet diseases. As a consequence, agonists and antagonists of the same AR subtype have been investigated for the treatment of similar pathologies. This approach is not paradoxical considering that the design of an effective therapy may depend on critical factors such as the state of disease progression, the dosage, the time of drug application, and the delivery method. Thus, AR agonists and antagonists, due to their potential, have been evaluated as drug candidates with many therapeutic applications [4, 76, 77, 182]. Since ARs are ubiquitous and involved in major physiological processes, biologically active compounds that interact with the adenosine system at different levels are still of interest.

3.6.1

Central Nervous System Disorders

Studies on adenosine and its receptors in the CNS are instrumental in understanding the pathogenic mechanism in many neurodegenerative disorders such as PD, Alzheimer’s (AD), Huntington’s disease, and ischemia. Furthermore, much evidence has highlighted that adenosine is also involved in pain [88, 89, 191]. In general, A1, A2A, and A3 ARs contribute to neuroprotection but are also involved in neurodegenerative processes. A2AAR is a promising therapeutic target for PD due to its interaction with the dopamine D2 receptor. The existence of hetero-receptor complexes (ARs-DRs), and the beneficial effect in PD of reciprocal antagonistic interaction [58] were demonstrated. Many A2AAR antagonists are under development and Istradefylline (Fig. 4), endowed with a safety profile and approved in combination with Levodopa for PD therapy, is commercially available in Japan. Furthermore, it was approved in 2019 even by the US Food and Drug Administration (FDA) for therapeutic use in PD [156]. Its approval process in Europe is still in progress [155, 156, 192]. The clinical evaluation of A2AAR antagonists, performed to test their efficacy in PD, also included Preladenant (Fig. 4) whose Phase III trial did not support evidence on its efficacy as monotherapy [193]. It has been repurposed to treat advanced solid tumors (also in combination with pembrolizumab), but the data did not support the study endpoints. Positive results after the clinical trial of another A2AAR antagonist, Tozadenant (Fig. 4), suggested its more promising profile with respect to

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

23

Preladenant [194]. However, the evaluation was discontinued and stopped due to serious side effects. A2AAR is also a therapeutic target for the development of AD and HD drugs. While in HD both A2A receptor agonists and antagonists have shown beneficial effects, leaving the question open [195], in AD selective A2AAR antagonists improve memory and can reduce brain β-amyloid (Aβ) levels [196]. It is worth noting that caffeine (Fig. 1), the most widely used drug, since contained in many consumer beverages, has been reported to reduce the risk of developing AD, leading to decreased brain Aβ formation, and prevent amyloid load and cognitive decline [197]. Furthermore, caffeine intake was associated with protective effects on cognitive impairment in both animals and humans [198, 199]. The health benefit of both coffee and tea consumption is related to folk medicine and experimental evidence [200]. However, caffeine is currently used as a CNS stimulant and in combination with analgesic [200] and has also been evaluated in clinical trials. The therapeutic use of AR ligands in cerebral ischemia was widely reported [201]. Although the roles of A3AR are controversial [202], the inhibitory effect of A1AR activation on excitotoxic glutamate release has been reported to lead to neuroprotection. Unfortunately, the use of selective A1AR agonists is hampered by peripheral side effects [68]. Several studies reported the neuroprotective mediatedactions of A3AR. In fact, as with A1AR, during ischemia, CA1 hippocampal A3 receptors affect neurotransmission leading to neuroprotection. However, the A3 subtype, depending on the situation, can also induce damage [202]. Some experimental evidence suggested that A1AR partial agonists could be used effectively as therapeutics in cerebral ischemia. Two CPA derivatives were reported to have in vitro neuroprotective effects after the application of oxygen-glucose deprivation (OGD) [203].

3.6.2

Pain

Many preclinical and clinical studies revealed that A1 and, to a lesser extent, A2AAR agonists were effective in counteracting pain. However, their therapeutic application has been hampered by adverse cardiovascular and motor side effects [89]. A1AR agonists have been reported to inhibit nociceptive input into the dorsal spinal cord [204] and to reduce pain in the periphery [2]. A2AAR agonists, though they exhibit some pronociceptive effects in the periphery, may be useful for inflammatory and neuropathic pain. Activation of the A2A receptor increases the release of interleukin-10 (IL-10) which is a potent antiinflammatory cytokine and leads to the suppression of pain. The A2AAR agonist Spongosine (2-methoxyadenosine, Fig. 3) isolated from a tropical marine sponge (Caribbean sponge Tectitethya crypta) demonstrated a diverse bioactivity profile that included anti-inflammatory activity and analgesic and vasodilation properties. It showed significant pain relief in an animal without producing typical side effects of AR agonists. Therefore, Spongosine was evaluated in a clinical trial for diabetic

24

D. Catarzi et al.

neuropathic pain, but the study was interrupted independently of the results obtained [205]. It is important to note that the role of A1 and A2A ARs in pain is still being debated because both agonists and antagonists have been reported to counteract pain of various origins [89, 206–211]. Although the A1 subtype has generally been reported to be a neuroprotective receptor [212, 213], some data suggest that prolonged stimulation of A1AR can lead to neurodegeneration. This result is due to the neuroprotective effects exerted by caffeine and other mixed A1/A2A AR antagonists, whose action has been attributed to the blockade of both A1 and A2A ARs [206, 214]. The potential of A2A receptor antagonists in treating pain was suggested [215] and successively confirmed [216, 218]. Clinical studies carried out to evaluate A1AR agonists for the treatment of chronic pain failed, although they showed efficacy in animal models [217]. Recurring evidence underlines that A3AR plays a role in pain [219–221]. In particular, A3AR agonists exerted remarkable and robust antinociceptive properties in acute and chronic pain while being devoid of side effects. This observation is supported by their good safety profile evidenced in non-pain clinical trials [222]. Some studies suggested that the use of A1AR partial agonists could be an alternative strategy to exploit the therapeutic potential of A1AR activation to treat pain. Adenosine-like compounds have been reported to show an antihyperalgesic effect comparable to that exerted by a full A1AR agonist as a reference compound. However, the reduction in pain was lower due to an improper pharmacokinetic profile [223].

3.6.3

Inflammation and Immune Diseases

Experimental evidence supports the use of antagonists and agonists of all ARs in the control of inflammatory and immune system responses [97, 224]. Since ARs are widely expressed on the surface of several immune cell families, the mediator adenosine can modulate their multiple functions, leading to maintenance of tissue integrity. However, excessive adenosine signaling can produce immune system suppression that promotes cancer development and sepsis progression [224]. This prolonged immune response together with dysregulation of immunomodulatory adenosine signaling can contribute to the development of autoimmune disorders. The natural alkaloid theophylline (Fig. 1) is currently used for the treatment of asthma. However, more potent and safer therapeutic agents have limited its use [225]. An overview of the roles of adenosine in modulation of the immune system with careful attention to potential adenosine-based therapies for autoimmune disorders was recently reported [226]. The review focused on the A3AR which can represent a very promising therapeutic target due to its overexpression in inflammatory cells. Another aspect that makes this AR subtype particularly attractive is that it is mainly activated in diseased tissues due to its up-regulation in autoimmune diseases [227].

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

25

Currently, IB-MECA (Fig. 3), a moderately selective A3AR agonist, is in a Phase III clinical trial for patients affected by psoriasis due to its demonstrated efficacy and safety profile [78, 228]. It has also been clinically evaluated for the treatment of rheumatoid arthritis, showing a significant anti-rheumatic effect. Its analog Cl-IBMECA (Fig. 3) is clinically evaluated for autoimmune inflammatory diseases [49, 78]. Even A2A and A2B ARs are involved in inflammation and both agonists and antagonists have been proposed as therapeutic agents [182]. A2AAR agonists, showing good outcomes in an animal model of asthma and chronic obstructive pulmonary disease (COPD), have been evaluated in affected patients. Unfortunately, the clinical studies failed [229]. The FDA-approved synthetic A2AAR agonist Regadenoson (Fig. 3) is currently clinically used as a diagnostic tool for myocardial perfusion imaging [230]. The approval for human treatment allows its evaluation in various clinical trials [77]. Regadenoson is currently being evaluated clinically in lung transplantation due to its efficacy to attenuate ischemia-reperfusion injury at this level [231]. Selective A2A agonists have been administered by inhalation to overcome severe cardiocirculatory effects in clinical trials to treat asthma. However, the trials failed due to their low efficacy [49].

3.6.4

Cancer

Adenosine levels increase in hypoxia that occurs in the solid tumor environment, thus affecting cancer growth. However, most ARs may have opposing roles in modulating the cell cycle or immunity, this depends on different factors [227, 232]. Interestingly, A3AR is overexpressed in several cancer cells and tissues and is therefore likely to have an important antitumor role [233]. Adenosine, through the A3AR subtype, activates the protection of tissues. In line with this assumption, many A3AR agonists showed their efficacy in different animal models of cancer [4, 98, 172, 234]. Two of these, i.e., IB-MECA and Cl-IB-MECA (Fig. 3), are currently in clinical trials for different cancer. In particular, Cl-IB-MECA, which shows both high safety and efficacy profiles, is in phase II trial for hepatocellular carcinoma [235]. The A3AR agonist IB-MECA was shown to be effective in an animal model of different types of metastatic carcinomas after oral administration [49]. Its synergistic action with cyclophosphamide in the murine lung metastatic melanoma model was demonstrated [97]. The high expression of the A3AR in many types of cancer cells, together with the growing experimental evidence accumulated over the years, makes it a promising target for drug development and also an effective tumor biomarker [184]. Other innovative tools can be the A2AAR antagonists as immune checkpoint blockers in oncology. Their effectiveness in counteracting cancer can be attributed to the ability to potentiate the immune system response [4, 184, 232, 236] and is supported by their safety profiles evidenced in clinical trials for PD. Several pharmaceutical companies have started clinical evaluations of A2AAR antagonists for

26

D. Catarzi et al.

oncology immunotherapy. As an example, the A2AAR antagonist Preladenant, whose phase 3 trial for PD failed, has been evaluated for the treatment of advanced solid tumors [78]. Several A2AAR antagonists have been evaluated, but their clinical study has failed, and some others are still in clinical trials in oncology. Recently, iTeos Therapeutics produced a report on the Phase 1 trial of the A2A receptor antagonist Inupadenant (EOS-850, 7-amino-10-[2-[4-[2,4-difluoro-5-[2-[(S)-methylsulfinyl] ethoxy]phenyl]piperazin-1-yl]ethyl]-4-(furan-2-yl)-12-thia-3,5,6,8,10pentazatricyclo[7.3.0.0]dodeca-1(9),2,4,7-tetraen-11-one) (Fig. 4) indicating its immunomodulatory and antitumor activity when administered in monotherapy [237]. It is reported to produce T-lymphocytes activation and proliferation and stimulation of a T-cell-mediated immune response against tumor cells. Inupadenant was generally well tolerated with the initial evidence of clinical benefit, consistent with previously reported safety data [238]. It is also orally active and does not penetrate brain. The company will continue to evaluate Inupadenant in the ongoing Phase 1b/2a trial in combination with a monoclonal antibody (Pembrolizumab) and chemotherapy, initially focusing on patients with prostate cancer, resistant melanoma, and breast cancer. Even A2BAR is highly expressed in tumor cells, representing a potential target for cancer therapy. In fact, some A2BAR antagonists are in clinical trials as antitumor agents [97, 98]. However, some evidence demonstrated that the roles of A2BAR in cancer are complex and sometimes controversial. Synergistic action of A2A/A2B AR antagonists was taken into consideration for the treatment of cancer. In fact, they are under clinical evaluation for a polypharmacological treatment in combination with a checkpoint inhibitor [236]. Concomitant AR blockade may improve the efficacy of the chemo/immunotherapy protocol. In particular, small-molecule dual A2A/A2B receptor antagonist, Etrumadenant (AB928, 3-[2-amino-6[1-[[6-(2-hydroxypropan-2-yl)pyridin-2-yl]methyl]triazol-4-yl]pyrimidin-4-yl]-2methylbenzonitrile) (Fig. 4), when tested in Phase 1 clinical trials showed good safety and pharmacokinetic profiles after oral administration [239]. More importantly, a significant decrease in tumor growth rate was observed when administered alone or better in combination with chemotherapy. The ongoing clinical trials include the evaluation of two potent and selective A2A receptor antagonists, Taminadenant (NIR178, PBF-509, 5-bromo-2,6-di (1H-pyrazol-1-yl)pyrimidin-4-amine) and Ciforadenant (Fig. 4). Taminadenant is a potent, no-furan, pyrimidine-based, and orally active antagonist developed by Novartis and Palobiofarma. It was endowed with a potential antineoplastic activity that could reactivate the antitumor immune response. Taminadenant has been introduced in clinical trials for many diseases such as PD and various types of cancer, in combination with a checkpoint inhibitor. In association with Spartalizumab (antiPD-1 Ab), it showed preliminary antitumor activity in a Phase 1b study in non-small cell lung carcinoma (NSCLC) [240]. The results of the Phase 2 study in patients with microsatellite stable (MSS) colorectal cancer (CRC) showed the efficacy of the therapeutic combination that was well tolerated, leading to clinical benefit [241].

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

27

Ciforadenant is an orally active A2AAR antagonist created by Vernalis. It was designed to address the chemical structural liabilities that may have led to concerns about toxicity of Vipadenant [168]. Ciforadenant blocks A2AAR expressed on the surface of immune cells (T-lymphocytes, natural killer (NK) cells, macrophages), thus abrogating adenosine-induced immunosuppression in the tumor microenvironment. This may stimulate antitumor immune responses leading to tumor regression. The preclinical evaluation of Ciforadenant in combination with other checkpoint inhibitors in solid tumors showed the potentiality of this therapeutic approach. Tumors were eliminated in up to 90% of treated mice. Thus, these results suggested the evaluation of Ciforadenant as a therapeutic agent in patients with solid tumors [242]. Ciforadenant was shown to be also active in other preclinical tumor models, both as monotherapy and in combination with anti-PD-L1 [243]. Different clinical studies on Ciforadenant started (safety and tolerability, against refractory multiple myeloma, solid tumors, and advanced cancers), most of which were completed but without posted results. A report from a Phase 1/1b clinical trial demonstrated that Ciforadenant was active alone and in combination with Atezolizumab (anti_PD-L1) in patients with advanced refractory renal cell cancer [244].

3.6.5

Cardiovascular and Related Diseases

Each AR has been taken into consideration as a target for the development of ligandbased therapies against a wide range of cardiovascular and related diseases. Adenosine itself is currently used for myocardial perfusion imaging for the diagnosis of coronary disease. Through the activation of A2A receptors, it causes coronary vasodilation with consequent variations in blood flow. However, fast adenosine metabolism prevents severe side effects such as cardiac block mediated by the A1 subtype. In contrast, acute bronchospasm is a recognized side effect related to activation of “off-target” A2BAR. Therefore, in recent times, Regadenoson [190, 191] (Fig. 3) has replaced adenosine for coronary stress imaging. It has been approved for use in humans both in the USA and in the European Union. Regadenoson is a short half-life selective A2A agonist that can be the best choice for patients with known bronchospasms [245]. In addition, the antiarrhythmic action of adenosine is exploited for the treatment of supraventricular tachycardia [246]. The scarcely selective A3 agonist IB-MECA (Fig. 3) was shown to be cardioprotective in cardiomyocyte progenitor cells [247]. This effect was ascribed to activation of both A3 and A2A ARs. Cl-IB-MECA also exerted protection against cardiotoxicity by reducing the inflammatory response and apoptosis [248]. Several A1AR ligands are currently in preclinical studies or are being developed for therapeutic applications. The non-nucleoside A1AR partial agonist, Capadenoson [249] (Fig. 3), was evaluated in a clinical trial in patients with angina [144], but this study was discontinued. In another phase II clinical study for the treatment of persistent or permanent atrial fibrillation, it did not show to effectively modulate heart rate [146]. However, an analog of Capadenoson, i.e., Neladenoson in the form of its prodrug (Neladenoson bialanate) [150] (Fig. 3), was shown to be safe

28

D. Catarzi et al.

for the treatment of chronic heart failure without cardiac or neurological side effects [151] but the clinical trial failed [78]. In fact, a lack of restorative effects on cardiac structure and function, and on exercise capacity, was reported, as well as a decrease in renal function [250, 251]. Cardiovascular activity of Capadenoson [10] must be attributed not only to activation of the A1AR but also the A2B subtype, which promotes cardioprotection and modulates cardiac fibrosis in the diseased heart [148, 252]. A2BAR activation is required for A1AR-mediated cardioprotection [253]. The greater effectiveness and safety of the Neladenoson bialanate prodrug with respect to Capadenoson is probably due to its higher A1AR selectivity leading to fewer undesirable side effects [151]. Interestingly, A1AR partial agonists have also been shown to be effective in reducing blood glucose levels by improving the insulin sensitivity [90, 254]. A2AAR agonists can be considered potential therapeutics in cardiovascularrelated diseases. In fact, some of them, in addition to being cardioprotective as a consequence of their anti-inflammatory effects, modulate platelet aggregation [255– 257] and cholesterol homeostasis [258] leading to the potential development of new cardiovascular therapies [259].

4 Perspectives Despite extensive research efforts from the pharmaceutical industry and academia, only a limited number of drugs targeting ARs have reached the market [78]. This is mainly due to the complexity of signaling, as well as the ubiquitous distribution of ARs in both healthy and diseased tissues. In fact, as a consequence, adenosine activates the protection of tissues and cells against injury, but it also stimulates various adverse effects, especially in the cardiovascular and respiratory systems [5]. Although the countless limitations still do not allow us to clarify the numerous dark aspects of adenosine and its receptors, it is undeniable to recognize the great progress that all researchers working in this field have contributed to making. Curiosity continues to fuel research on this topic, and a glance at the current publications gives a measure of the ever-new interest. Over the years, many important issues have been solved and, at the same time, many important new aspects, up to now ignored or underestimated, have emerged as essential for the development of new drugs. What follows is just a brief mention of the critical issues that have emerged and the new frontiers to be set as objectives for the years to come. The importance of ligand-binding kinetics in the drug discovery process has been underestimated. Although the affinity of a ligand is important for predicting its therapeutic potential, all kinetic characteristics can highly influence the possibility that a biologically active compound could be developed as a drug. More attention should be paid to this concept to reduce the failures of future clinical trials. The availability of the residence time of orthosteric ligands may allow the choice of a candidate compound with the appropriate residence times for a specific therapeutic

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

29

use [78]. Furthermore, kinetic studies can also shed light on the mechanism of AR allosteric modulators. These latter potentially represent a novel therapeutic strategy for some pathological conditions as an alternative to orthosteric ligands that often lack sufficient on-target selectivity thus showing adverse effects [128, 182, 260]. The allosteric modulator can stabilize active and/or inactive receptor conformation(s), thus influencing the kinetics of a ligand bound within the orthosteric site, in addition to its affinity and/or efficacy. The ability of an AR allosteric ligand to modulate adenosine signaling predominantly in damaged tissues and organs, thus allowing spatiotemporal control, results in a reduction in adverse effects [127, 261]. Allosteric modulators can selectively elicit a physiological response only where and when the orthosteric ligand is released. Modulation of kinetic characteristics has been useful in generating compounds with an “infinite” residence time since their bond to the target persists over time. These are covalent ligands, i.e., compounds that irreversibly bind to the receptor because they are endowed with a reactive moiety versus specific amino acid residues of the binding site. Covalent ligands as well as many other kinds of AR probes gave a contribution to clarifying the structural biology of ARs that suffer from numerous limitations [62]. For example, the structure of human A1AR was elucidated by X-ray crystallography with the covalent antagonist, DU172 (4-[3-(8-cyclohexyl-2,6-dioxo1-propyl-7H-purin-3-yl)propylcarbamoyl]benzenesulfonyl fluoride) [117] (Fig. 4). Over the past decades, a diverse array of molecular probes that can be used to investigate receptor structure and function have proven invaluable in AR research. However, though advanced techniques have generated a great number of structures for GPCRs including A1 and A2A ARs, structure elucidation of A2B and A3 receptors has not yet been successful. These will be the challenges that experts in this field will face in the coming years. Moreover, the lack of experimental techniques capable of evaluating dynamics opens the question of characterization of the thermodynamic and kinetic properties of ligand binding to ARs [262]. An additional focus has to be placed on the particular phenomenon of some AR ligands, biased agonism, also known as functional selectivity. Exploiting the ability of a receptor to differentially activate therapeutic signaling pathways while avoiding others that could lead to side effects appears to be an intriguing strategy. Biased AR agonists, which can preferentially signal through a G protein-independent pathway, can be considered innovative smart drugs potentially endowed with fewer side effects. This new aspect of AR signaling should be taken into account for the design of new drugs, to reduce the possibility of clinical trial failure [104]. Although different AR ligands have been studied at the preclinical level to fight cancer, a few molecules have progressed to clinical trials. The A3AR agonist Cl-IBMECA is under clinical evaluation for the treatment of advanced hepatocellular carcinoma treatment. However, the A3AR appears to have a dual nature, as evidence of pro-tumoral and antitumoral activity suggests that other mechanisms are involved in the anticancer properties of A3AR ligands [227, 232]. Therefore, an in-depth

30

D. Catarzi et al.

investigation must be performed taking into account also that extrapolation from preclinical animal studies to humans is hazardous due to the different expression and functions of A3AR in the various animal species [106]. Several A2AAR antagonists are being evaluated. In fact, A2AAR has recently received renewed attention for its roles in immuno-oncology [263]. Although early clinical studies have shown modest benefits with A2AAR blockade, considerable work is needed to identify new polypharmacological strategies in which checkpoint inhibition is associated with other immune-based therapies [264]. In addition to A2AAR, the A2B subtype was also proposed as a novel target for the (immuno)therapy of cancer. In fact, the discovery of new molecules capable of antagonizing A2BAR or both A2B and A2A subtypes opened a new perspective in pharmacotherapy. However, only a few A2BAR antagonists have reached clinical trials [78]. Currently, the most likely scenario is that AR modulation could serve as an additional therapy to other anticancer strategies (chemo-, immune- and radio-therapy) to combat a wide variety of cancers. A polypharmacological approach has a higher probability of success than a one-target therapy to treat not only cancer but also many other diseases with high social impact [265]. The multifactorial character of these diseases suggests exploiting the simultaneous modulation of multiple targets involved in the disorder that must be fought. Thus, a new goal may be the discovery of multitarget-directed ligands (MTDLs) (two or more birds with one stone) to provide a strong synergistic effect against a specific pathology. MTDLs can be designed by combining two (or more) different pharmacophoric features by using diverse strategies leading, in general, to bitopic, bivalent, and dual-acting ligands [266, 267]. The advantages of a ligand obtained with the merging approach over ligands designed by pharmacophoric linkage or fusion (reduction of molecular weight, topological polar surface area, and clogP) could result in improved “drug-like” properties. The MTDL approach resulted from the observation that compounds modulating a single target might not be optimal drug candidates for the treatment of complex diseases [268]. Thus, to overcome this issue, a single molecule with dual activity could be a more effective drug because it is endowed with higher efficacy, better pharmacokinetic profiles, and also compliance with the patient. This strategy is expected to have many advantages compared to the coadministration of different drugs. Over the years, some progress has been made. Interesting ligands targeting one or more ARs, or one AR and other GPCRs (such as β-adrenergic, dopaminergic, histaminergic), or an enzyme (monoamine oxidase (MAO), carbonic anhydrases (CAs)) have been obtained and studied. Some of the more recent and/or interesting reports are suggested [269– 274]. Although few general reviews on the peculiarities of these types of ligands are available [266, 267], a detailed overview of progress in the field of MTDLs targeting ARs is needed.

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

31

5 Concluding Remarks In conclusion, the rapid progress of studies on adenosine and its receptors has had a strong impact on the drug discovery process. Much evidence indicated that ARs are involved in the physiological regulation of several homeostatic processes and, as a consequence, in the etiology of different pathologies. Moreover, understanding the molecular biology of each AR has provided a solid basis for considering them as targets for adenosine-based pharmacotherapies. Previously and newly reported discoveries allowed the identification of increasingly active and selective AR ligands endowed with a safety profile. Thus, the therapeutic potential of these AR ligands was evidenced and the frontiers for their admission to clinical trial programs were opened. Perspectives for compounds endowed with adequate kinetic properties or that interfere with adenosine signaling are very encouraging. Emerging as potential innovative therapeutic agents, they are likely to reach the role of drugs for clinical use. Attention should be paid to A2AAR antagonists whose innovative mechanism of action in oncology has been validated. In fact, the development of A2AAR antagonists as anticancer agents seems to rapidly progress, and some of them are already being evaluated in a clinical trial. This attractive therapeutic use is highly competitive and is supported by the safety profile of these compounds that was evidenced in clinical trials for PD. Similar consideration can be given to A3AR agonists that have emerged not only as promising anticancer agents but also as successful drugs to counteract pain. Therefore, they are or are being tested in clinical trials for neuropathic pain [275]. As in the case of A2AAR antagonists, their good safety profile was evidenced in non-pain clinical studies, thus supporting their potential role as therapeutics in humans. Last but not least, it is important to underline the well-known efficacy of A1AR partial agonists in cardiovascular diseases, such as chronic heart failure, atrial fibrillation, and angina, as documented by the results obtained in clinical trials. However, the anti-inflammatory effect that makes them effective in cardiovascular-related diseases could work in synergy to develop new cardiovascular therapies. From a future perspective, polypharmacology seems to represent an innovative therapeutic strategy [265]. In particular, small “drug-like” molecules targeting two different receptor binding sites simultaneously could be the best choice. The development of MTDLs has become a promising approach in drug design and discovery to overcome various problems related to the administered drugs and/or therapeutic protocols. In fact, it remains one of the major challenges in drug development and opens new avenues for rational design of the next generation of more effective but less toxic therapeutic agents. The search for novel AR-targeted MTDLs with adequate balanced activities could represent a new strategy to combat multifactorial-dependent diseases in which adenosine is involved. The goal of developing AR ligands as drugs accepted

32

D. Catarzi et al.

for clinical use in humans appears to be getting closer and closer. This is what every researcher working in this field continues to do with the tenacity to try to achieve with a contribution, be it small or large. Compliance with Ethical Standards Conflict of Interest The author declares that they have no conflict of interest. Funding Original research of our team is funded by the Italian Ministry for University and Research (MIUR, PRIN 2017MT3993_004 project). Ethical Approval This chapter does not contain any studies with human participants or animals performed by the authors.

References 1. Williams RA (1989) Purine receptors. In: Williams M, Glennon RA, Timmermans PBMWM (eds) Receptor pharmacology and function. Weiner M, Dekker M, New York, pp 503–525 2. Borea PA, Gessi S, Merighi S, Varani K (2016) Adenosine as a multi-signalling guardian angel in human diseases: when, where and how does it exert its protective effects? Trends Pharmacol Sci 37:419–434 3. Cunha RA (2001) Adenosine as a neuromodulator and as a homeostatic regulator in the nervous system: different roles, different sources and different receptors. Neurochem Int 38: 107–125 4. Borea PA, Varani K, Gessi S, Merighi S, Vincenzi F (2019) The adenosine receptors. In: Di Giovanni G (ed) The receptors, vol 34. Humana Press, pp 1–593 5. Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K (2017) Pathological overproduction: the bad side of adenosine. Br J Pharmacol 174:1945–1960 6. Scheele KW (1776) Analysis of the calculus vesicae. Essay IX. In: The chemical essays of Charles-William Scheele. Translated from the transactions of the Academy of Sciences at Stockholm, with additions (1786). J. Murray, London, pp 199–214 7. Fischer E (1890) Synthesen in der Zuckergruppe. Ber Deut Chem Ges 23:2114–2141 8. Fischer E (1899) Synthesen in der Puringruppe. Ber Deut Chem Ges 32:435–504 9. Fischer E (1907) Untersuchungen in Der Puringruppe: (1882–1906). Springer Verlag, Berlin 10. Kossel A (1885) Ueber eine neue Base aus dem Tierkorper. Ber Deut Chem Ges 18:79–81 11. Kossel A (1888) Ueber eine neue Base aus dem Pflanzenreich. Ber Deut Chem Ges 21:2164– 2167 12. Levene PA, Jacobs WA (1911) Ueber die Inosinsaure. Vierte Mitteilung Ber Deut Chem Ges 44:746–752 13. Maruyama K (1991) The discovery of adenosine triphosphate and the establishment of its structure. J Hist Biol 24:145–154 14. Drury AN, Szent-Gyorgyi A (1929) The physiological activity of adenine compounds with especial reference to their action on the mammalian heart. J Physiol 68:213–237 15. Bennett DW, Drury AN (1931) Further observations relating to the physiological activity of adenine compounds. J Physiol 72:28–320 16. Gillespie JH (1934) The biological significance of the linkages in adenosine triphosphoric acid. J Physiol 80:345–359 17. Honey RM, Ritchie WT, Thomson WAR (1930) The action of adenosine upon the human heart. QJM os-23(92):485–489 18. Berne RM (1963) Cardiac nucleotides in hypoxia: possible role in regulation of coronary blood flow. Am J Physiol 204:317–322

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

33

19. Gerlach E, Deuticke B, Dreisbach RH (1963) Die Nucleotid-Abbauim Herzmuskel bei Sauerstoffmangel und seine mogliche Bedeutung fur die Coronardurchblutung. Naturwissenschaft 20. Burnstock G (1972) Purinergic nerves. Pharmacol Rev 24:509–581 21. Rubio R, Berne RM (1969) Release of adenosine by the normal myocardium in dogs and its relationship to the regulation of coronary resistance. Circ Res 25:407–415 22. Olsson RA (1970) Changes in content of purine nucleoside in canine myocardium during coronary occlusion. Circ Res 26:301–306 23. Fox AC, Reed GE, Glassman E, Kaltman AJ, Silk BB (1974) Release of adenosine from human hearts during angina induced by rapid atrial pacing. J Clin Invest 53:1447–1457 24. Sattin A, Rall TW (1970) The effect of adenosine and adenine nucleotides on the cyclic adenosine 3′,5′-phosphate content of Guinea pig cerebral cortex slices. Mol Pharmacol 6:13– 23 25. Shimizu H, Daly J (1970) Formation of cyclic adenosine 3′,5′-monophosphate from adenosine in brain slices. Biochim Biophys Acta 222(2):465–473 26. Bretschneider HJ, Frank A, Bernard U, Kochsiek K, Scheler F (1959) Effect of a pyrimidopyrimidine derivative on the oxygen supply of the heart muscle. Arzneimittelforschung 9:49– 59 27. Pfleger K, Seifen E, Schöndorf H (1969) Inosine potentiation of the effect of adenosine on the heart. Biochem Pharmacol 18:43–51 28. Dietmann K, Schaumann W, Thiel M (1981) Die Geschichte der Adenosin-Derivate. Ein Abenteuer der Arzniemittelforschung. Getbooks GmbH, Bad Camberg, Germany 29. Rubio R, Berne RM (1980) Location of purine and pyrimidine nucleoside phosphorylases in heart, kidney and liver. Am J Physiol 239:H721–H730 30. Hershfield MS, Kredich NM (1978) S-adenosylhomocysteine hydrolase is an adenosine binding protein: a target for adenosine toxicity. Science 202:757–760 31. Schrader J, Nees S, Gerlach E (1981) Role of S-adenosylhomocysteine hydrolase in adenosine metabolism in mammalian heart. Biochem J 196:65–70 32. Nagy JI, La Bella LA, Buss M, Daddona PE (1984) Immunohistochemistry of adenosine deaminase: implications for adenosine neurotransmission. Science 224(4645):166–168 33. Burnstock G (1978) A basis for distinguishing two types of purinergic receptor. In: Straub RW, Boils L (eds) Cell membrane receptors for drugs and hormones: a multidisciplinary approach. Raven Press, New York, pp 107–118 34. Fredholm BB (1982) Adenosine receptors. Med Biol 60(6):289–293 35. Van Calker D, Müller M, Hamprecht B (1979) Adenosine regulates via two different types of receptors, the accumulation of cyclic AMP in cultured brain cells. J Neurochem 33:999–1005 36. Londos C, Wolff J (1977) Two distinct adenosine-sensitive sites on adenylate cyclase. Proc Natl Acad Sci U S A 74:5482–5486 37. Schrader J, Baumann G, Gerlach E (1977) Adenosine as inhibitor of myocardial effects of catecholamines. Pflugers Arch Eur J Physiol 372:29–35 38. Böhm M, Brückner R, Neumann J, Schmitz W, Scholz H, Starbatty J (1986) Role of guanine nucleotide-binding protein in the regulation by adenosine of cardiac potassium conductance and force of contraction. Evaluation with pertussis toxin. Naunyn Schmiedebergs Arch Pharmacol 332:403–405 39. Dolphin AC, Forda SR, Scott RH (1986) Calcium-dependent currents in cultured rat dorsal root ganglion neurones are inhibited by an adenosine analogue. J Physiol 373:47–61 40. Bruns RF, Lu GH, Pugsley TA (1986) Characterization of the A2 adenosine receptor labeled by [3H]NECA in rat striatal membranes. Mol Pharmacol 29:331–346 41. Fredholm BB (1977) Activation of adenylate cyclase from rat striatum and tuberculum olfactorium by adenosine. Med Biol 55:262–267 42. Premont J, Perez M, Bockaert J (1977) Adenosine-sensitive adenylate cyclase in rat striatal homogenates and its relationship to dopamine- and Ca2+-sensitive adenylate cyclases. Mol Pharmacol 13:662–670

34

D. Catarzi et al.

43. Daly JW, Butts-Lamb P, Padgett W (1983) Subclasses of adenosine receptors in the central nervous system: interaction with caffeine and related methylxanthines. Cell Mol Neurobiol 3: 69–80 44. Ribeiro JA, Sebastião AM (1986) Adenosine receptors and calcium: basis for proposing a third (A3) adenosine receptor. Prog Neurobiol 26:179–209 45. Ali H, Cunha-Melo JR, Saul WF, Beaven MA (1990) Activation of phospholipase C via adenosine receptors provides synergisti signals for secretion in antigen-stimulated RBL-2H3 cells. Evidence for a novel adenosine receptor. J Biol Chem 265:745–753 46. Zhou QY, Li C, Olah ME, Johnson RA, Stiles GL, Civelli O (1992) Molecular cloning and characterization of an adenosine receptor: the A3 adenosine receptor. Proc Natl Acad Sci U S A 89:7432–7436 47. Fredholm BB, Ijzerman AP, Jacobson KA, Klotz KN, Linden J (2001) International Union of Pharmacology. XXV. Nomenclature and classification of adenosine receptors. Pharmacol Rev 53:527–552 48. Fredholm BB, IJzerman AP, Jacobson KA, Linden J, Müller CE (2011) International union of basic and clinical pharmacology. LXXXI. Nomenclature and classification of adenosine receptors – an update. Pharmacol Rev 63:1–34 49. Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K (2018) Pharmacology of adenosine receptors: the state of the art. Physiol Rev 98:1591–1625 50. Malbon CC, Hert RC, Fain JN (1978) Characterization of [3H]adenosine binding to fat cell membranes. J Biol Chem 253:3114–3122 51. Schwabe U, Trost T (1980) Characterization of adenosine receptors in rat brain by (-)[3H]N6phenylisopropyladenosine. Naunyn Schmiedebergs Arch Pharmacol 313:179–187 52. Williams M, Risley EA (1980) Biochemical characterization of putative central purinergic receptors by using 2-chloro[3H]adenosine, a stable analog of adenosine. Proc Natl Acad Sci U S A 77:6892–6896 53. Bruns RF, Fergus JH, Badger EW, Bristol JA, Santay LA, Hartman JD, Hays SJ, Huang CC (1987) Binding of the A1-selective adenosine antagonist 8-cyclopentyl-1,3-dipropylxanthine to rat membranes. Naunyn Schmied Arch Pharmacol 335:59–63 54. Jarvis MF, Williams M (1989) Direct autoradiographic localization of adenosine A2 receptors in the rat brain using the A2-selective agonist, [3H]CGS 21680. Eur J Pharmacol 168:243–246 55. Zocchi C, Ongini E, Conti A, Monopoli A, Negretti A, Baraldi PG, Dionisotti S (1996) The non-xanthine heterocyclic compound SCH 58261 is a new potent and selective A2a adenosine receptor antagonist. J Pharmacol Exp Ther 276(2):398–404 56. Poucher SM, Keddie JR, Singh P, Stoggall SM, Caulkett PW, Jones G, Coll MG (1995) The in vitro pharmacology of ZM 241385, a potent, non-xanthine A2a selective adenosine receptor antagonist. Br J Pharmacol 115:1096–1102 57. Olah ME, Gallo-Rodriguez C, Jacobson KA, Stiles GL (1994) 125I-4-aminobenzyl-5X-N methylcarboxamidoadenosine, a high affinity radioligand for the rat A3 adenosine receptor. Mol Pharmacol 45:978–982 58. Baraldi PG, Cacciari B, Romagnoli R, Varani K, Merighi S, Gessi S, Borea PA, Leung E, Hickey SL, Spalluto G (2000) Synthesis and preliminary biological evaluation of [3H]-MRE 3008-F20: the first high affinity radioligand antagonist for the human A3 adenosine receptors. Bioorg Med Chem Lett 10:209–211 59. Hinz S, Alnouri WM, Pleiss U, Müller CE (2018) Tritium-labeled agonists as tools for studying adenosine A2B receptors. Purinergic Signal 14:223–233 60. Federico S, Lassiani L, Spalluto G (2019) Chemical probes for the adenosine receptors. Pharmaceuticals 12:168 61. Yang X, Heitman LH, IJzerman AP, van der Es D (2021) Molecular probes for the human adenosine receptors. Purinergic Signal 17(1):85–108 62. Weichert D, Gmeiner P (2015) Covalent molecular probes for class A G protein-coupled receptors: advances and applications. ACS Chem Biol 10:1376–1386

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

35

63. Klotz KN (2000) Adenosine receptors and their ligands. Naunyn Schmiedebergs Arch Pharmacol 362(4–5):382–391 64. Ukena D, Padgett WL, Hong O, Daly JW, Daly DT, Olsson RA (1987) N6-substituted 9-methyladenines: a new class of adenosine receptor antagonists. FEBS Lett 215(2):203–208 65. Francis JE, Cash WD, Psychoyos S, Ghai G, Wenk P, Friedmann RC, Atkins C, Warren V, Furness P (1988) Structure-activity profile of a series of novel triazoloquinazoline adenosine antagonists. J Med Chem 31(5):1014–1020 66. Kiesman WF, Elzein E, Zablocki J (2009) A1 adenosine receptor antagonists, agonists, and allosteric enhancers. In: Wilson C, Mustafa S (eds) Adenosine receptors in health and disease. Handbook of experimental pharmacology, vol 193. Springer, Berlin, pp 25–58 67. Cristalli G, Müller CE, Volpini R (2009) Recent developments in adenosine A2A receptor ligands. In: Wilson CN, Mustafa SJ (eds) Adenosine receptors in health and disease. Handbook of experimental pharmacology, vol 193. Springer, Berlin, pp 59–98 68. Deb PK, Deka S, Borah P, Abed SN, Klotz K-N (2019) Medicinal chemistry and therapeutic potential of agonists, antagonists and allosteric modulators of A1 adenosine receptor: current status and perspectives. Curr Pharm Des 25:2697–2715 69. Preti D, Baraldi PG, Moorman AR, Borea PA, Varani K (2015) History and perspectives of A2A adenosine receptor antagonists as potential therapeutic agents. Med Res Rev 35:790–848 70. Guerrero A (2018) A2A adenosine receptor agonists and their potential therapeutic applications. An update. Curr Med Chem 25:3597–3612 71. Kalla RV, Zablocki J, Tabrizi MA, Baraldi PG (2009) Recent developments in A2B adenosine receptor ligands. In: Wilson C, Mustafa S (eds) Adenosine receptors in health and disease. Handbook of experimental pharmacology, vol 193. Springer, Berlin, pp 99–122 72. Jacobson KA, Klutz A, Tosh DK, Ivanov AA, Preti D, Baraldi PG (2009) Medicinal chemistry of the A3 adenosine receptor: agonists, antagonists, and receptor engineering. In: Wilson C, Mustafa S (eds) Adenosine receptors in health and disease. Handbook of experimental pharmacology, vol 193. Springer, Berlin, pp 123–159 73. Müller CE, Jacobson KA (2011) Recent developments in adenosine receptor ligands and their potential as novel drugs. Biochim Biophys Acta 1808:1290–1308 74. Gao Z-G, Jacobson KA (2011) Emerging adenosine receptor agonists-an update. Expert Opin Emerg Drugs 16(4):597–602 75. Dal Ben D, Lambertucci C, Buccioni M, Navia AM, Marucci G, Spinaci A, Volpini R (2019) Non-nucleoside agonists of the adenosine receptors: an overview. Pharmaceuticals 12:150 76. Saini A, Patel R, Gaba S, Singh G, Gupta GD, Monga V (2022) Adenosine receptor antagonists: recent advances and therapeutic perspective. Eur J Med Chem 227:113907 77. Jacobson KA, Tosh DK, Jain S, Gao Z-G (2019) Historical and current adenosine receptor agonists in preclinical and clinical development. Front Cell Neurosci 13:124 78. IJzerman AP, Jacobson KA, Müller CE, Cronstein BN, Cunha RA (2022) International Union of Basic and Clinical Pharmacology. CXII: adenosine receptors: a further update. Pharmacol Rev 74:340–372 79. Di Marco JP, Sellers TD, Berne RM, West GA, Belardinelli L (1985) Diagnostic and therapeutic use of adenosine in patients with supraventricular tachyarrhythmias. J Am Coll Cardiol 6:417–425 80. Verani M (1991) Pharmacological stress with adenosine for myocardial perfusion imaging. Semin Nucl Med 21:266–272 81. Newby AC (1984) Adenosine and the concept of “retaliatory metabolites”. Trends Biochem Sci 9:42–44 82. Zimmermann H (2000) Extracellular metabolism of ATP and other nucleotides. Naunyn Schmiedebergs Arch Pharmacol 362:299–309 83. Chen JF, Eltzschig HK, Fredholm BB (2013) Adenosine receptors as drug targets – what are the challenges? Nat Rev Drug Discov 12:265–286

36

D. Catarzi et al.

84. Deussen A, Stappert M, Schäfer S, Kelm M (1999) Quantification of extracellular and intracellular adenosine production: understanding the transmembranous concentration gradient. Circulation 99:2041–2047 85. Peleli M, Fredholm BB, Sobrevia L, Carlstrӧm M (2017) Pharmacological targeting of adenosine receptor signaling. Mol Aspects Med 55:4–8 86. Gracia E, Farré D, Cortés A, Ferrer-Costa C, Orozco M, Mallol J, Lluís C, Canela EI, McCormick PJ, Franco R, Fanelli F, Casadó V (2013) The catalytic site structural gate of adenosine deaminase allosterically modulates ligand binding to adenosine receptors. FASEB J 27:1048–1061 87. Pacheco R, Martinez-Navio JM, Lejeune M, Climent N, Oliva H, Gatell JM, Gallart T, Mallol J, Lluis C, Franco R (2005) CD26, adenosine deaminase, and adenosine receptors mediate costimulatory signals in the immunological synapse. Proc Natl Acad Sci U S A 102: 9583–9588 88. Liu Y-J, Chen J, Li X, Zhou X, Hu Y-M, Chu S-F, Peng Y, Chen N-H (2019) Research progress on adenosine in central nervous system diseases. CNS Neurosci Ther 25:899–910 89. Sawynok J (2016) Adenosine receptor targets for pain. Neuroscience 338:1–18 90. Dhalla AK, Chisholm JW, Reaven GM, Belardinelli L (2009) A1 adenosine receptor: role in diabetes and obesity. Handb Exp Pharmacol 193:271–295 91. Varani K, Vincenzi F, Merighi S, Gessi S, Borea PA (2017) Biochemical and pharmacological role of A1 adenosine receptors and their modulation as novel therapeutic strategy. Adv Exp Med Biol 1051:193–232 92. Rabadi MM, Lee HT (2015) Adenosine receptors and renal ischaemia reperfusion injury. Acta Physiol 213:222–231 93. Hua X, Erikson CJ, Chason KD, Rosebrock CN, Deshpande DA, Penn RB, Tilley SL (2007) Involvement of A1 adenosine receptors and neural pathways in adenosine-induced bronchoconstriction in mice. Am J Physiol Lung Cell Mol Physiol 293:L25–L32 94. Wilson CN, Nadeem A, Spina D, Brown R, Page CP, Mustafa SJ (2009) Adenosine receptors and asthma. Handb Exp Pharmacol 193:329–362 95. Wolska N, Rozalski M (2019) Blood platelet adenosine receptors as potential targets for antiplatelet therapy. Int J Mol Sci 20(21):5475 96. Janes K, Esposito E, Doyle T, Cuzzocrea S, Tosh DK, Jacobson KA, Salvemini D (2014) A3 adenosine receptor agonist prevents the development of paclitaxel-induced neuropathic pain by modulating spinal glial-restricted redox-dependent signaling pathways. Pain 155:2560– 2567 97. Jacobson KA, Merighi S, Varani K, Borea PA, Baraldi S, Aghazadeh Tabrizi M, Romagnoli R, Baraldi PA, Ciancetta A, Tosh DK, Gao Z-G, Gessi S (2018) A3 adenosine receptors as modulators of inflammation: from medicinal chemistry to therapy. Med Res Rev 38(4):1031–1072 98. Marucci G, Santinelli C, Buccioni M, Martí Navia A, Lambertucci C, Zhurina A, Yli-Harjab O, Volpini R, Kandhavelu M (2018) Anticancer activity study of A3 adenosine receptor agonists. Life Sci 205:155–163 99. Hussain A, Gharanei AM, Nagra AS, Maddock HL (2014) Caspase inhibition via A3 adenosine receptors: a new cardioprotective mechanism against myocardial infarction. Cardiovasc Drugs Ther 28:19–32 100. Gao Z-G, Inoue A, Jacobson KA (2018) On the G protein-coupling selectivity of the native A2B adenosine receptor. Biochem Pharmacol 151:201–213 101. Cordeaux Y, IJzerman AP, Hill SJ (2004) Coupling of the human A1 adenosine receptor to different heterotrimeric G proteins: evidence for agonist-specific G protein activation. Br J Pharmacol 143:705–714 102. Kull B, Svenningsson P, Fredholm BB (2000) Adenosine A2A receptors are colocalized with and activate golf in rat striatum. Mol Pharmacol 58(4):771–777

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

37

103. Schulte G, Fredholm BB (2000) Human adenosine A(1), A(2A), A(2B), and A(3) receptors expressed in Chinese hamster ovary cells all mediate the phosphorylation of extracellularregulated kinase 1/2. Mol Pharmacol 58:477–482 104. McNeill SM, Jo-Anne B, White PJ, May Lauren T (2021) Biased agonism at adenosine receptors. Cell Signal 82:109954 105. Wall MJ, Hill E, Huckstepp R, Barkan K, Deganutti G, Leuenberger M, Preti B, Winfield I, Wei H, Imlach W et al (2020) A biased adenosine A1R agonist elicits analgesia without cardiorespiratory depression. bioRxiv. 2020.04.04.023945 106. Alnouri MW, Jepards S, Casari A, Schiedel AC, Hinz S, Müller CE (2015) Selectivity is species-dependent: characterization of standard agonists and antagonists at human, rat, and mouse adenosine receptors. Purinergic Signal 11:389–407 107. Du L, Gao ZG, Paoletta S, Wan TC, Gizewski ET, Barbour S, van Veldhoven JPD, IJzerman AP, Jacobson KA, Auchampach JA (2018) Species differences and mechanism of action of A3 adenosine receptor allosteric modulators. Purinergic Signal 14:59–71 108. Fredholm BB, Arslan G, Halldner L et al (2000) Structure and function of adenosine receptors and their genes. Naunyn Schmiedebergs Arch Pharmacol 362:364–374 109. Jaakola V-P, Griffith MT, Hanson MA, Cherezov V, Chien EYT, Lane JR, Ijzerman AP, Stevens RC (2008) The 2.6 angstrom crystal structure of a human A2A adenosine receptor bound to an antagonist. Science 322:1211–1217 110. Doré AS, Robertson N, Errey JC, Ng I, Hollenstein K, Tehan B, Hurrell E, Bennett K, Congreve M, Magnani F, Tate CG, Weir M, Marshall FH (2011) Structure of the adenosine A(2A) receptor in complex with ZM241385 and the xanthines XAC and caffeine. Structure 19:1283–1293 111. Lebon G, Warne T, Edwards PC, Bennett K, Langmead CJ, Leslie AGW, Tate CG (2011) Agonist-bound adenosine A2A receptor structures reveal common features of GPCR activation. Nature 474:521–525 112. Xu F, Wu H, Katritch V, Han GW, Jacobson KA, Gao Z-G, Cherezov V, Stevens RC (2011) Structure of an agonist-bound human A2A adenosine receptor. Science 332:322–327 113. Carpenter B, Nehmé R, Warne T, Leslie AG, Tate CG (2016) Structure of the adenosine A (2A) receptor bound to an engineered G protein. Nature 536:104–107 114. Barrington WW, Jacobson KA, Stiles GL (1989) Demonstration of distinct agonist and antagonist conformations of the A1 adenosine receptor. J Biol Chem 264:13157–13164 115. Olah ME, Jacobson KA, Stiles GL (1994) Role of the second extracellular loop of adenosine receptors in agonist and antagonist binding: analysis of chimeric A1/A3 adenosine receptors. J Biol Chem 269:24692–24698 116. Tosh DK, Paoletta S, Deflorian F et al (2012) Structural sweet spot for A1 adenosine receptor activation by truncated (N)-methanocarba nucleosides: receptor docking and potent anticonvulsant activity. J Med Chem 55:8075–8090 117. Glukhova A, Thal DM, Nguyen AT, Vecchio EA, Jörg M, Scammells PJ, May LT, Sexton PM, Christopoulos A (2017) Structure of the adenosine A1 receptor reveals the basis for subtype selectivity. Cell 168:867–877 118. Cheng Y (2018) Single-particle cryo-EM-how did it get here and where will it go. Science 361: 876–880 119. Ceska T, Chung CW, Cooke R, Phillips C, Williams PA (2019) Cryo-EM in drug discovery. Biochem Soc Trans 47:281–293 120. Draper-Joyce CJ, Khoshouei M, Thal DM, Liang YL, Nguyen ATN, Furness SGB, Venugopal H, Baltos JA, Plitzko JM, Danev R (2018) Structure of the adenosine-bound human adenosine A1 receptor-Gi complex. Nature 558:559–563 121. García-Nafría J, Lee Y, Bai X, Carpente B, Tate CG (2018) Cryo-EM structure of the adenosine A2A receptor coupled to an engineered heterotrimeric G protein. Elife 7:e35946 122. Wang J, Bhattarai A, Ahmad WI, Farnan TS, John KP, Miao Y (2020) Computer-aided GPCR drug discovery. In: Jastrzebska B, Park PSH (eds) GPCRs. Academic Press, Cambridge, pp 283–293

38

D. Catarzi et al.

123. Sussman JL, Lin D, Jiang J, Manning NO, Prilusky J, Ritter O, Abola EE (1998) Protein Data Bank (PDB): database of three-dimensional structural information of biological macromolecules. Acta Crystallogr Sect D Biol Crystallogr 54:1078–1084 124. Karplus M, Mc Cammon JA (2002) Molecular dynamics simulations of biomolecules. Nat Struct Mol Biol 9:646–652 125. Wang J, Miao Y (2019) Mechanistic insights into specific G protein interactions with adenosine receptors. J Phys Chem B 123(30):6462–6473 126. Wang J, Bhattarai A, Do HN, Akhter S, Miao Y (2022) Molecular simulations and drug discovery of adenosine receptors. Molecules 27:2054 127. May LT, Leach K, Sexton PM, Christopoulos A (2007) Allosteric modulation of G proteincoupled receptors. Annu Rev Pharmacol Toxicol 47:1–51 128. Vecchio EA, Baltos J-A, Nguyen ATN, Christopoulos A, White PJ, May LT (2018) New paradigms in adenosine receptor pharmacology: allostery, oligomerization and biased agonism. Br J Pharmacol 175(21):4036–4046 129. Korkutata M, Agrawal L, Lazarus M (2022) Allosteric modulation of adenosine A2A receptors as a new therapeutic avenue. Int J Mol Sci 23(4):2101 130. Poulsen S-A, Quinn RJ (1998) Adenosine receptors: new opportunities for future drugs. Bioorg Med Chem 6(6):619–641 131. Wu PH, Phillis JW (1982) Adenosine receptor in rat brain membranes: characterization of high affinity binding of [3H]-2-chloroadenosine. Int J Biochem 14:399–404 132. Müller CE, Baqi Y, Hinz S, Namasivayam V (2018) Medicinal chemistry of A2B adenosine receptors. In: Borea PA, Varani K, Gessi S, Merighi S, Vincenzi F (eds) The adenosine receptors. The receptors, vol 34. Springer, Cham, pp 137–168 133. Beukers MW, Chang LCW, von Frijtag Drabbe Kunzel JK, Mulder-Krieger T, Spanjersberg RF, Brussee J, IJzermann AP (2004) New, non-adenosine, high-potency agonists for the human adenosine A2B receptor with an improved selectivity profile compared to the reference agonist N-ethylcarboxamidoadenosine. J Med Chem 47:3707–3709 134. Hinz S, Lacher SK, Seibt BF, Müller CE (2014) BAY60-6583 acts as a partial agonist at adenosine A2B receptors. J Pharmacol Exp Ther 349:427–436 135. Guo M, Gao ZG, Tyler R, Stodden T, Li Y, Ramsey J, Zhao WJ, Wang GJ, Wiers CE, Fowler JS, Rice KC, Jacobson KA, Kim SW, Volkow ND (2018) Preclinical evaluation of the first adenosine A1 receptor partial agonist radioligand for positron emission tomography imaging. J Med Chem 61:9966–9975 136. Bertarelli DC, Diekmann M, Hayallah AM, Rüsing D, Iqbal J, Preiss B, Verspohl EJ, Müller CE (2006) Characterization of human and rodent native and recombinant adenosine A (2B) receptors by radioligand binding studies. Purinergic Signal 2:559–571 137. Betti M, Catarzi D, Varano F, Falsini M, Varani K, Vincenzi F, Dal Ben D, Lambertucci C, Colotta V (2018) The aminopyridine-3,5-dicarbonitrile core for the design of new nonnucleoside-like agonists of the human adenosine A2B receptor. Eur J Med Chem 150:127–139 138. Johnston-Cox H, Koupenova M, Yang D, Corkey B, Gokce N, Farb MG, LeBrasseur N, Ravid K (2012) The A2B adenosine receptor modulates glucose homeostasis and obesity. PloS One 7: e40584 139. Johnston-Cox H, Eisenstein AS, Koupenova M, Carroll S, Ravid K (2014) The macrophage A2B adenosine receptor regulates tissue insulin sensitivity. PloS One 9:e98775 140. Csoka B, Koscso B, Toro G, Kokai E, Virag L, Nemeth ZH, Pacher P, Bai P, Hasko G (2014) A2B adenosine receptors prevent insulin resistance by inhibiting adipose tissue inflammation via maintaining alternative macrophage activation. Diabetes 63:850–866 141. Tian Y, Piras BA, Kron IL, French BA, Yang Z (2015) Adenosine 2B receptor activation reduces myocardial reperfusion injury by promoting anti-inflammatory macrophages differentiation via PI3K/Akt pathway. Oxid Med Cell Longev 2015:585297 142. Ni Y, Liang D, Tian Y, Kron IL, French BA, Yang Z (2018) Infarct-sparing effect of adenosine A2B receptor agonist is primarily due to its action on splenic leukocytes via a PI3K/Akt/IL-10 pathway. J Surg Res 232:442–449

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

39

143. Bott-Flugel L, Bernshausen A, Schneider H, Luppa P, Zimmermann K, Albrecht-Kupper B, Kast R, Laugwitz KL, Ehmke H, Knorr A, Seyfarth M (2011) Selective attenuation of norepinephrine release and stress-induced heart rate increase by partial adenosine A1 agonism. PloS One 6(3):e18048 144. Tendera M, Gaszewska-Zurek E, Parma Z, Ponikowski P, Jankowska E, Kawecka-Jaszcz K, Czarnecka D, Krzeminska-Pakula M, Bednarkiewicz Z, Sosnowski M, Kilama MO, Agrawal R (2012) The new oral adenosine A1 receptor agonist capadenoson in male patients with stable angina. Clin Res Cardiol 101:585–591 145. Sabbah HN, Gupta RC, Kohli S, Wang RS, Zhang K, Zimmermann K, Diedrichs N, AlbrechtKupper BE (2013) Chronic therapy with a partial adenosine A1-receptor agonist improves left ventricular function and remodeling in dogs with advanced heart failure. Circ Heart Fail 6: 563–571 146. Albrecht-Kupper BE, Leineweber K, Nell PG (2012) Partial adenosine A1 receptor agonists for cardiovascular therapies. Purinergic Signal 8:91–99 147. Bailey IR, Laughlin B, Moore LA, Bogren LK, Barati Z (2017) Drew KL optimization of thermolytic response to A1 adenosine receptor agonists in rats. J Pharm Exp Ther 362:424–430 148. Baltos JA, Vecchio EA, Harris MA, Qin CX, Ritchie RH, Christopoulos A, White PJ, May LT (2017) Capadenoson, a clinically trialed partial adenosine A1 receptor agonist, can stimulate adenosine A2B receptor biased agonism. Biochem Pharmacol 135:79–89 149. Rueda P, Merlin J, Chimenti S, Feletou M, Paysant J, White PJ, Christopoulos A, Sexton PM, Summers RJ, Charman WN, May LT, Langmead CJ (2021) Pharmacological insights into safety and efficacy determinants for the development of adenosine receptor biased agonists in the treatment of heart failure. Front Pharmacol 12:628060 150. Meibom D, Albrecht-Kupper B, Diedrich N, Hubsch W, Kast R, Kramer T, Krenz U, Lerchen HG, Mittendorf J, Nell PG, Süssmeier F, Vakalopoulos A, Zimmermann K (2017) Neladenoson bialanate hydrochloride: a prodrug of a partial adenosine A1 receptor agonist for the chronic treatment of heart diseases. Chem Med Chem 12:728–737 151. Voors AA, Dungen HD, Senni M, Nodari S, Agostoni P, Ponikowski P, Bax JJ, Butler J, Kim RJ, Dorhout B, Dinh W, Gheorghiade M (2017) Safety and tolerability of neladenoson bialanate, a novel oral partial adenosine A1 receptor agonist, in patients with chronic heart failure. J Clin Pharmacol 57:440–451 152. Louvel J, Guo D, Soethoudt M, Mocking TA, Lenselink EB, Mulder-Krieger T, Heitman LH, IJzerman AP (2015) Structure-kinetics relationships of Capadenoson derivatives as adenosine A1 receptor agonists. Eur J Med Chem 101:681–691 153. Fredholm BB (1985) On the mechanism of action of theophylline and caffeine. Acta Med Scand 217:149–153 154. Müller CE, Jacobson KA (2011) Xanthines as adenosine receptor antagonists. Handb Exp Pharmacol 200:151–199 155. Takahashi M, Fujita M, Asai N, Saki M, Mori A (2018) Safety and effectiveness of istradefylline in patients with Parkinson’s disease: interim analysis of a postmarketing surveillance study in Japan. Expert Opin Pharmacother 19:1635–1642 156. Chen J-F, Cunha RA (2020) The belated US FDA approval of the adenosine A2A receptor antagonist istradefylline for treatment of Parkinson’s disease. Purinergic Signal 16(2):167–174 157. Borrmann T, Hinz S, Bertarelli DCG, Li W, Florin NC, Scheiff AB, Müller CE (2009) 1-Alkyl-8-(piperazine-1-sulfonyl)phenylxanthines: development and characterization of adenosine A2B receptor antagonists and a new radioligand with subnanomolar affinity and subtype specificity. J Med Chem 52(13):3994–4006 158. Ji X, Kim YC, Ahern DG, Linden J, Jacobson KA (2001) [3H]MRS 1754, a selective antagonist radioligand for A2B adenosine receptors. Biochem Pharmacol 61:657–663 159. Rohilla S, Bansal R, Kachler S, Klotz KN (2019) Synthesis, biological evaluation and molecular modelling studies of 1, 3, 7, 8-tetrasubstituted xanthines as potent and selective A2A AR ligands with in vivo efficacy against animal model of Parkinson's disease. Bioorg Chem 87:601–612

40

D. Catarzi et al.

160. Koul S, Ramdas V, Barawkar DA, Waman YB, Prasad N, Madadi SK, Shejul YD, Bonagiri R, Basu S, Menon S, Reddy SB (2017) Design and synthesis of novel, potent and selective hypoxanthine analogs as adenosine A1 receptor antagonists and their biological evaluation. Bioorg Med Chem 25:1963–1975 161. Basu S, Barawkar DA, Ramdas V, Waman Y, Patel M, Panmand A, Kumar S, Thorat S, Bonagiri R, Jadhav D, Mukhopadhyay P (2017) A2B adenosine receptor antagonists: design, synthesis and biological evaluation of novel xanthine derivatives. Eur J Med Chem 127:986– 996 162. Basu S, Barawkar DA, Ramdas V, Patel M, Waman Y, Panmand A, Kumar S, Thorat S, Naykodi M, Goswami A, Reddy BS (2017) Design and synthesis of novel xanthine derivatives as potent and selective A2B adenosine receptor antagonists for the treatment of chronic inflammatory airway diseases. Eur J Med Chem 134:218–229 163. de Lera RM, Lim Y-H, Zeng J (2014) Adenosine A2A receptor as a drug discovery target. J Med Chem 57:3623–3650 164. Gatta F, Del Giudice MR, Borioni A, Borea PA, Dionisotti S, Ongini E (1993) Synthesis of imidazo[1,2-c]pyrazolo[4,3-e]pyrimidines, pyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidines and 1,2,4- triazolo[5,1-i]purines: new potent adenosine A2 receptor antagonists. Eur J Med Chem 28:569–576 165. Baraldi PG, Tabrizi MA, Fruttarolo F, Bovero A, Avitabile B, Preti D, Romagnoli R, Merighi S, Gessi S, Varani K, Borea PA (2003) Recent developments in the field of A3 adenosine receptor antagonists. Drug Dev Res 58:315–329 166. Le Witt PA, Aradi SD, Hauser RA, Rascol O (2020) The challenge of developing adenosine A2A antagonists for Parkinson disease: istradefylline, preladenant, and tozadenant. Parkinsonism Relat Disord 80(Suppl 1):S54–S63 167. Gillespie RJ, Cliffe IA, Dawson C, Dourish CT, Gaur S, Jordan AM, Knight AR, Lerpiniere J, Misra A, Pratt RM, Roffey J, Stratton GC, Upton R, Weiss SM, Williamson DS (2008) Antagonists of the human adenosine A2A receptor. Part 3: design and synthesis of pyrazolo [3,4-d]pyrimidines, pyrrolo[2,3-d]-pyrimidines and 6-arylpurines. Bioorg Med Chem Lett 18(9):2924–2929 168. Gillespie RJ, Bamford SJ, Botting R, Come M, Denny S, Gaur S, Griffin M, Jordan AM, Knight AR, Lerpiniere J, Leonardi S, Lightowler S, Mc Ateer S, Merrett A, Misra A, Padfield A, Reece M, Saadi M, Selwood DL, Stratton GC, Surry D, Todd R, Tong X, Ruston V, Upton R, Weiss SM (2009) Antagonist of the human A2A adenosine receptor. 4. Design, synthesis and preclinical evaluations of 7-aryltriazolo[4,5-d]pyrimidines. J Med Chem 52(1):33–47 169. Basu S, Barawkar DA, Thorat S, Shejul YD, Patel M, Naykodi M, Jain V, Salve Y, Prasad V, Chaudhary S, Ghosh I (2017) Design, synthesis of novel, potent, selective, orally bioavailable adenosine A2A receptor antagonists and their biological evaluation. J Med Chem 60:681–694 170. Renk DR, Skraban M, Bier D, Schulze A, Wabbals E, Wedekind F, Neumaier F, Neumaier B, Holschbach M (2021) Design, synthesis and biological evaluation of Tozadenant analogues as adenosine A2A receptor ligands. Eur J Med Chem 15(214):113214 171. Li A-N, Moro S, Forsyth N, Melman N, Ji X-D, Jacobson KA (1999) Synthesis, CoMFA analysis, and receptor docking of 3,5-diacyl-2,4-dialkylpyridine derivatives as selective A3 adenosine receptor antagonists. J Med Chem 42:706–721 172. Madi L, Bar-Yehuda S, Barer F, Ardon E, Ochaion A, Fishman P (2003) A3 adenosine receptor activation in melanoma cells: association between receptor fate and tumor growth inhibition. J Biol Chem 278(43):42121–42130 173. Moro S, Gao ZG, Jacobson KA, Spalluto G (2006) Progress in pursuit of therapeutic adenosine receptor antagonists. Med Res Rev 26:131–159 174. Jacobson KA, IJzerman AP, Müller CE (2021) Medicinal chemistry of P2 and adenosine receptors: common scaffolds adapted for multiple targets. Biochem Pharmacol 187:114311

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

41

175. Cheng RKY, Segala E, Robertson N, Deflorian F, Doré AS, Errey JC, Fiez-Vandal C, Marshall FH, Cooke RM (2017) Structures of human A1 and A2A adenosine receptors xanthines reveal determinants of selectivity. Structure 25:1275–1285 176. Jespers W, Schiedel AC, Heitman LH, Cooke R, Gutiérrez-de-Terán H (2018) Structural mapping of adenosine receptor mutations: ligand binding and signaling mechanisms. Trends Pharmacol Sci 39:75–89 177. Carpenter B, Lebon G (2017) Human adenosine A2A receptor: molecular mechanism of ligand binding and activation. Front Pharmacol 8:898 178. Rucktooa P, Cheng RKY, Segala E, Geng T, Errey JC, Brown GA, Cooke RM, Marshall FH, Doré AS (2018) Towards high throughput GPCR crystallography: in Meso soaking of adenosine A2A receptor crystals. Sci Rep 8:41 179. Costanzi S, Ivanov AA, Tikhonova IG, Jacobson KA (2007) Structure and function of G protein-coupled receptors studied using sequence analysis, molecular modeling and receptor engineering: adenosine receptors. Front Drug Des Discov 3:63–79 180. Dal Ben D, Lambertucci C, Marucci G, Volpini R, Cristalli G (2010) Adenosine receptor modeling: what does the A2A crystal structure tell us? Curr Top Med Chem 10:993–1018 181. Ciancetta A, Jacobson KA (2017) Structural probing and molecular modeling of the A3 adenosine receptor: a focus on agonist binding. Molecules 22:449 182. Effendi WI, Nagano T, Kobayashi K, Nishimura Y (2020) Focusing on adenosine receptors as a potential targeted therapy in human diseases. Cell 9:785 183. Sousa JB, Fresco P, Diniz C, Goncalves J (2018) Adenosine receptor ligands on cancer therapy. A review of patent literature. Recent Pat Anticancer Drug Discov 13:40–69 184. Merighi S, Battistello E, Giacomelli L, Varani K, Vincenzi F, Borea PA (2019) Targeting A3 and A2A adenosine receptors in the fight against cancer. Exp Opin Ther Targets 23:669–678 185. Yu F, Zhu C, Xie Q, Wang Y (2020) Adenosine A2A receptor antagonists for cancer immunotherapy. J Med Chem 63:12196–12212 186. Guglielmi P, Carradori S, Campestre C, Poce G (2019) Novel therapies for glaucoma: a patent review (2013-2019). Exp Opin Ther Patents 29:769–780 187. Zheng J, Zhang X, Zhen X (2019) Development of adenosine A2A receptor antagonists for the treatment of Parkinson’s disease: a recent update and challenge. ACS Chem Nerosci 10:783– 791 188. Borah P, Deka S, Mailavaram RP, Deb PK (2019) P1 receptor agonists/antagonists in clinical trials – potential drug candidates of the future. Curr Pharm Des 25:2792–2807 189. Chandrasekaran B, Samarneh S, Jaber AMY, Kassab G, Agrawal N (2019) Therapeutic potentials of A2B adenosine receptor ligands: current status and perspectives. Curr Pharm Des 25:2741–2771 190. Iskandrian AE, Bateman TM, Belardinelli L, Blackburn B, Cerqueira MD, Hendel RC, Lieu H, Mahmarian JJ, Olmsted A, Underwood SR, Vitola J, Wang W (2007) Adenosine versus regadenoson comparative evaluation in myocardial perfusion imaging: results of the ADVANCE phase 3 multicenter international trial. J Nucl Cardiol 14(5):645–658 191. Stone TW, Ceruti S, Abbracchio MP (2009) Adenosine receptors and neurological disease: neuroprotection and neurodegeneration. In: Wilson CN, Mustafa SJ (eds) Adenosine receptors in health and disease, vol 193. Springer, Berlin, pp 535–587 192. Jenner P, Mori A, Aradi SD, Hauser RA (2021) Istradefylline – a first generation adenosine A2A antagonist for the treatment of Parkinson’s disease. Expert Rev Neurother 21:317–333 193. Stocchi F, Rascol O, Hauser RA, Huyck S, Tzontcheva A, Capece R, Ho TW, Sklar P, Lines C, Michelson D, Hewitt DJ (2017) Randomized trial of preladenant, given as monotherapy, in patients with early Parkinson disease. Neurology 88:2198–2206 194. Hauser RA, Olanow CW, Kieburtz KD, Pourcher E, Docu-Axelerad A, Lew M, Kozyolkin O, Neale A, Resburg C, Meya U, Kenney C, Bandak S (2014) Tozadenant (SYN115) in patients with Parkinson’s disease who have motor fluctuations on levodopa: a phase 2b, double-blind, randomised trial. Lancet Neurol 13:767–776

42

D. Catarzi et al.

195. Popoli P, Blum D, Domenici M, Burnouf S, Chern YA (2008) Critical evaluation of adenosine A2A receptors as potentially “Druggable” targets in Huntington’s disease. Curr Pharm Des 14: 1500–1511 196. Faivre E, Coelho JE, Zornbach K, Malik E, Baqi Y, Schneider M, Cellai L, Carvalho K, Sebda S, Figeac M, Eddarkaoui S, Caillierez R, Chern Y, Heneka M, Sergeant N, Müller CE, Halle A, Buée L, Lopes LV, Blum D (2018) Beneficial effect of a selective adenosine A2A receptor antagonist in the APPswe/PS1dE9 mouse model of Alzheimer’s disease. Front Mol Neurosci 11:235 197. Eskelinen MH, Kivipelto M (2010) Caffeine as a protective factor in dementia and Alzheimer’s disease. J Alzheimers Dis 20:S167–S174 198. Santos C, Lunet N, Azevedo A, de Mendonça A, Ritchie K, Barros H (2010) Caffeine intake is associated with a lower risk of cognitive decline: a cohort study from Portugal. J Alzheimers Dis 20(Suppl 1):S175–S185 199. Chen JQA, Scheltens P, Groot C, Ossenkoppele R (2020) Associations between caffeine consumption, cognitive decline, and dementia: a systematic review. J Alzheimers Dis 78(4): 1519–1546 200. van Dam RM, Hu FB, Willett WC (2020) Coffee, caffeine, and health. N Engl J Med 383:369– 378 201. Melani A, Pugliese AM, Pedata F (2014) Chapter thirteen-adenosine receptors in cerebral ischemia. Int Rev Neurobiol 119:309–348 202. Pedata F, Pugliese AM, Coppi E, Popoli P, Morelli M, Schwarzschild MA, Melani A (2007) Adenosine in the central nervous system: effects on neurotransmission and neuroprotection. Immun Endoc Metab Agents Med Chem 7:304–321 203. Martire A, Lambertucci C, Pepponi R, Ferrante A, Benati N, Buccioni M, Dal Ben D, Marucci G, Klotz K-N, Volpini R, Popoli P (2019) Neuroprotective potential of adenosine A1 receptor partial agonists in experimental models of cerebral ischemia. J Neurochem 149: 211–230 204. Dickenson AH, Suzuki R, Reeve AJ (2000) Adenosine as a potential analgesic target in inflammatory and neuropathic pains. CNS Drugs 13:77–85 205. Knezevic NN, Cicmil N, Knezevic I, Candido KD (2015) Discontinued neuropathic pain therapy between 2009-2015. Expert Opin Investig Drugs 24:1631–1646 206. Rivera-Oliver M, Díaz-Ríos M (2014) Using caffeine and other adenosine receptor antagonists and agonists as therapeutic tools against neurodegenerative diseases: a review. Life Sci 101:1– 9 207. Bilkei-Gorzo A, Abo-Salem OM, Hayallah AM, Michel K, Müller CE, Zimmer A (2008) Adenosine receptor subtype-selective antagonists in inflammation and hyperalgesia. Naunyn Schmiedebergs Arch Pharmacol 377:65–76 208. Pechlivanov DM, Georgiev VP (2005) Effects of single and long-term theophylline treatment on the threshold of mechanical nociception: contribution of adenosine A1 and α2adrenoceptors. Methods Find Exp Clin Pharmacol 27:659–664 209. Gong Q-J, Li Y-Y, Xin W-J, Wei X-H, Cui Y, Wang J, Liu Y, Liu C-C, Li Y-Y (2010) Differential effects of adenosine A1 receptor on pain-related behavior in normal and nerveinjured rats. Brain Res 1361:23–30 210. Katz NK, Ryals JM, Wright DE (2015) Central or peripheral delivery of an adenosine A1 receptor agonist improves mechanical allodynia in mouse model of painful diabetic neuropathy. Neuroscience 285:312–323 211. Ferre S, Diamond I, Goldberg SR, Yao L, Hourani SMO, Huang ZL, Urade Y, Kitchen I (2007) Adenosine A2A receptor in ventral striatum, hypothalamus and nociceptive circuitry. Implication for drug addiction, sleep and pain. Prog Neurobiol 83:332–347 212. Cunha RA (2005) Neuroprotection by adenosine in the brain: from A1 receptor activation to A2A receptor blockade. Purinergic Signal 1:111–134 213. Stockwell J, Jakova E, Cayabyab FS (2017) Adenosine A1 and A2A receptors in the brain: current research and their role in neurodegeneration. Molecules 22(4):676

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

43

214. Betti M, Catarzi D, Varano F, Falsini M, Varani K, Vincenzi F, Pasquini S, Di Cesare ML, Ghelardini C, Lucarini E, Dal Ben D, Spinaci A, Bartolucci G, Menicatti M, Colotta V (2019) Modifications on the Amino-3,5-dicyanopyridine core to obtain multifaceted adenosine receptor ligands with antineuropathic activity. J Med Chem 62:6894–6912 215. Ledent C, Vaugeois JM, Shiffmann SN, Pedrazzini T, El Yacoubini M, Vanderhaeghen JJ, Costentin J, Heath JK, Vassert G, Parmentier M (1997) Aggressiveness, hypoalgesia and high blood pressure in mice lacking the adenosine A2A receptor. Nature 388:647–678 216. Falsini M, Catarzi D, Varano F, Ceni C, Dal Ben D, Marucci G, Buccioni M, Volpini R, Di Cesare ML, Lucarini E, Ghelardini C, Bartolucci G, Menicatti M, Colotta V (2019) Antioxidant-conjugated 1,2,4-triazolo[4,3-a]pyrazin-3-one derivatives: highly potent and selective human A2A adenosine receptor antagonists possessing protective efficacy in neuropathic. J Med Chem 62:8511–8531 217. Imlach WL, Bhola RF, May LT, Christopoulos A, Christie MJ (2015) A positive allosteric modulator of the A1 AR selectively inhibits primary afferent synaptic transmission in a neuropathic pain model. Mol Pharmacol 88:460–468 218. Varano F, Catarzi D, Vincenzi F, Betti M, Falsini M, Ravani A, Borea PA, Colotta V, Varani K (2016) Design, synthesis, and pharmacological characterization of 2-(2-furanyl)thiazolo [5,4-d]pyrimidine-5,7-diamine derivatives: new highly potent A2A adenosine receptor inverse agonists with antinociceptive activity. J Med Chem 59:10564–10576 219. Salvemini D, Jacobson KA (2017) Highly selective A3 adenosine receptor agonists relieve chronic neuropathic pain. Expert Opin Ther Patents 27:967 220. Janes K, Symons-Liguori AM, Jacobson KA, Salvemini D (2016) Identification of A3 adenosine receptor agonists as novel non-narcotic analgesics. Br J Pharmacol 173:1253–1267 221. Terayama R, Tabata M, Maruhama K, Iida S (2018) A3 adenosine receptor agonist attenuates neuropathic pain by suppressing activation of microglia and convergence of nociceptive inputs in the spinal dorsal horn. Exp Brain Res 236:3203–3213 222. Van Troostenburg AR, Clark EV, Carey WDH, Warrington SJ, Kerns WD, Cohn I, Silverman MH, Bar-Yehuda S, Fong K-LL, Fishman P (2004) Tolerability, pharmacokinetics and concentration-dependent hemodynamic effects of oral CF101, an A3 adenosine receptor agonist, in healthy young men. Int J Clin Pharmacol Ther 42:534–542 223. Schaddelee MP, Collins SD, DeJongh J, de Boer AG, Ijzerman AP, Danhof M (2005) Pharmacokinetic/pharmacodynamic modelling of the anti-hyperalgesic and anti-nociceptive effect of adenosine A1 receptor partial agonists in neuropathic pain. Eur J Pharmacol 514:131– 140 224. Antonioli L, Fornai M, Blandizzi C, Pacher P, Haskó G (2019) Adenosine signaling and the immune system: when a lot could be too much. Immunol Lett 205:9–15 225. Journey JD, Bentley TP (2020) Theophylline toxicity. In: StatPearls, StatPearls Publishing LLC, Treasure Island. https://www.ncbi.nlm.nih.gov/books/NBK532962/ 226. Magni G, Ceruti S (2020) Adenosine signaling in autoimmune disorders. Pharmaceuticals 13: 260 227. Gessi S, Merighi S, Varani K, Leung E, Mac Lennan S, Borea PA (2008) The A3 adenosine receptor: an enigmatic player in cell biology. Pharmacol Ther 117:123–140 228. David M, Akerman L, Ziv M, Kadurina M, Gospodinov D, Pavlotsky F, Yankova R, Kouzeva V, Ramon M, Silverman MH, Fishman P (2012) Treatment of plaque-type psoriasis with oral CF101: data from an exploratory randomized phase 2 clinical trial. J Eur Acad Dermatol Venereol 26:361–367 229. Mantell S, Jones R, Trevethick M (2010) Design and application of locally delivered agonists of the adenosine A2A receptor. Expert Rev Clin Pharmacol 3:55–72 230. Palani G, Ananthasubramaniam K (2013) Regadenoson: review of its established role in myocardial perfusion imaging and emerging applications. Cardiol Rev 21:42–48 231. Sharma AK, La Par DJ, Stone ML, Zhao Y, Mehta CK, Kron IL, Laubach VE (2016) NOX2 activation of natural killer T cells is blocked by the adenosine A2A receptor to inhibit lung ischemia-reperfusion injury. Am J Respir Crit Care Med 193:988–999

44

D. Catarzi et al.

232. Gessi S, Merighi S, Sacchetto V, Simioni C, Borea PA (2011) Adenosine receptors and cancer. Biochim Biophys Acta 1808:1400–1412 233. Ren T, Tian T, Feng X, Ye S, Wang H, Wu W, Qiu Y, Yu C, He Y, Zeng J, Cen J, Zhou Y (2015) An adenosine A3 receptor agonist inhibits DSS-induced colitis in mice through modulation of the NF-κB signaling pathway. Sci Rep 5:9047 234. Ohana G, Bar-Yehuda S, Arich A, Madi L, Dreznick Z, Rath-Wolfson L, Silberman D, Slosman G, Fishman P (2003) Inhibition of primary colon carcinoma growth and liver metastasis by the A3 adenosine receptor agonist CF101. Br J Cancer 89:1552–1558 235. Stemmer SM, Manojlovic NS, Vasile Marinca M, Petrov P, Cherciu N, Ganea D, Ciuleanu T-E, Puscas IA, Shaalan Beg M, Purcell WT, Croitoru A-E, Nedyalkova Ilieva R, Natošević S, Lavinir Nita A, Kalev DN, Harpaz Z, Farbstein M, Silverman MH, Fishman P, Llovet JM (2019) A phase II, randomized, double-blind, placebo-controlled trial evaluating efficacy and safety of namodenoson (CF102), an A3 adenosine receptor agonist (A3AR), as a second-line treatment in patients with child-Pugh B (CPB) advanced hepatocellular carcinoma (HCC). J Clin Oncol 37:2503–2503 236. Congreve M, Brown GA, Borodovsky A, Lamb ML (2018) Targeting adenosine A2A receptor antagonism for treatment of cancer. Expert Opin Drug Discovery 13:997–1003 237. Buisseret L, Rottey S, De Bono JS, Migeotte A, Delafontaine B, Manickavasagar T, Martinoli C, Wald N, Rossetti M, Gangolli EA, Wiegert E, Ni MI, Lager JJ, Machiels J-PH (2021) Phase 1 trial of the adenosine A2A receptor antagonist inupadenant (EOS-850): update on tolerability, and antitumor activity potentially associated with the expression of the A2A receptor within the tumor. J Clin Oncol 39(15):2562–2562 238. Houthuys E, Brouwer M, Nyawouame F, Pirson R, Marillier R, Deregnaucourt T, Marchante J, Swiercz J, Moulin C, Bol V, Driessens G, Detheux M, Quéva C, Crosignani S, Gomes B (2017) A novel adenosine A2A receptor antagonist optimized for high potency in adenosinerich tumor microenvironment boost antitumor immunity. Cancer Res 77(13):1683 239. Seitz L, Jin L, Leleti M, Ashok D, Jeffrey J, Rieger A, Tiessen RG, Arold G, Tan JBL, Powers JP, Walters MJ, Karakunnel J (2019) Safety, tolerability, and pharmacology of AB928, a novel dual adenosine receptor antagonist, in a randomized, phase 1 study in healthy volunteers. Invest New Drugs 37:711–721 240. Chiappori AA, Creelan B, Tanvetyanon T, Gray JE, Haura EB, Thapa R, Barlow L, Chen Z, Chen DT, Beg AA, Boyle TA, Castro J, Morgan L, Morris E, Aregay M, FeK H, Manenti L, Antonia S (2022) Phase I study of taminadenant (PBF509/NIR178), an A2A receptor antagonist, with or without spartalizumab (PDR001), in patients with advanced non-small cell lung cancer. Clin Cancer Res 28(11):2313–2320 241. Saavedra Santa Gadea O, Greil R, de Jonge MJA, Tan D, Jerusalem G, Damian S, Grell P, Wainberg ZA, Wolf J, Joerger M, Carlino MS, Kasper S, Yap TA, Otero J, Yang X, Nesbitt V, Kim J, Ho Lee L, Choudhury S, Leal TA (2021) Phase (Ph) II study of taminadenant (NIR178) + spartalizumab (PDR001) in patients (pts) with microsatellite stable (MSS) colorectal cancer (CRC). Ann Oncol 32(5):S553 242. Willingham SB, Ho Po Y, Hotson A, Hill C, Piccione EC, Hsieh J, Liu L, Buggy JJ, McCaffery I, Miller RA (2018) A2AR antagonism with CPI-444 induces antitumor responses and augments efficacy to anti-PD-(L)1 and anti-CTLA-4 in preclinical models. Cancer Immunol Res 6(10):1136–1149 243. Leone RD, Sun I-M, Oh M-H, Sun I-H, Wen J, Englert J, Powell JD (2018) Inhibition of the adenosine A2a receptor modulates expression of T cell coinhibitory receptors and improves effector function for enhanced checkpoint blockade and ACT in murine cancer models. Cancer Immunol Immunother 67(8):1271–1284 244. Fong HA, Powderly JD, Sznol M, Heist RS, Choueiri TK, George S, Hughes BGM, Hellmann MD, Shepard DR, Rini BI, Kummar S, Weise AM, Riese MJ, Markman B, Emens LA, Mahadevan D, Luke JJ, Laport G, Brody JD, Hernandez-Aya L, Bonomi P, Goldman JW, Berim L, Renouf DJ, Goodwin RA, Munneke B, Ho PY, Hsieh J, McCaffery I, Kwei L,

Once Upon a Time Adenosine and Its Receptors: Historical Survey. . .

45

Willingham SB, Miller RA (2020) Adenosine A2A receptor blockade as an immunotherapy for treatment-refractory renal cell cancer. Cancer Discov 10(1):40–53 245. Patel JJ, Alzahrani T (2021) Myocardial perfusion scan. In: StatPearls, StatPearls Publishing LLC, Treasure Island. https://www.ncbi.nlm.nih.gov/books/NBK539772/ 246. Singh S, McKintosh R (2022) Adenosine. In: StatPearls. StatPearls Publishing LLC, Treasure Island. https://www.ncbi.nlm.nih.gov/books/NBK519049/ 247. Tian Y, Marshall M, French BA, Linden J, Yang Z (2015) The infarct-sparing effect of IB-MECA against myocardial ischemia/reperfusion injury in mice is mediated by sequential activation of adenosine A3 and A2A receptors. Basic Res Cardiol 110:16 248. Galal A, El-Bakly WM, Al Haleem ENA, El-Demerdash E (2016) Selective A3 adenosine receptor agonist protects against doxorubicin-induced cardiotoxicity. Cancer Chemother Pharmacol 77:309–322 249. Rosentreter U, Krämer T, Shimada M, Hübsch W, Diedrichs N, Krahn T, Henninger K, Stasch J-P, Wischnat R (2003) Substituted 2-thio-3,5-dicyano-4-phenyl-6-aminopyridines and the use of the same. WO 2003053441 A1 250. Voors AA, Bax JJ, Hernandez AF, Wirtz AB, Pap AF, Ferreira AC, Senni M, van der Laan M, Butler J (2019) Safety and efficacy of the partial adenosine A1 receptor agonist neladenoson bialanate in patients with chronic heart failure with reduced ejection fraction: a phase IIb, randomized, double-blind, placebo-controlled trial. Eur J Heart Fail 21:1426–1433 251. Shah S.J, Voors AA, McMurray JJV, Kitzman DW, Viethen T, Bomfim Wirtz A, Huang E, Pap AF, Solomon SD (2012) Effect of neladenoson bialanate on exercise capacity among patients with heart failure with preserved ejection fraction: a randomized clinical trial. JAMA 321:2101–2112 252. Lasley RD (2018) Adenosine receptor-mediated cardioprotection-current limitations and future directions. Front Pharmacol 9:310 253. Zhan E, McIntosh VJ, Lasley RD (2011) Adenosine A2A and A2B receptors are both required for adenosine A1 receptor-mediated cardioprotection. Am J Physiol Heart Circ Physiol 301: H1183–H1189 254. Dhalla AK, Santikul M, Smith M, Wong M-Y, Shryock JC, Belardinelli L (2007) Antilipolytic activity of a novel partial A1 adenosine receptor agonist devoid of cardiovascular effects: comparison with nicotinic acid. J Pharmacol Exp Ther 321(1):327–333 255. Fuentes E, Fuentes M, Caballero J, Palomo I, Hinz S, El-Tayeb A, Müller CE (2018) Adenosine A2A receptor agonists with potent antiplatelet activity. Platelets 29:292–300 256. Wolska N, Kassassir NH, Luzak B, Watala C, Rozalski M (2020) Adenosine receptor agonists increase the inhibition of platelet function by P2Y12 antagonists in a cAMP and calciumdependent manner. Pharmaceuticals 13:177 257. Johnston-Cox HA, Ravid K (2011) Adenosine and blood platelets. Purinergic Signal 7:357– 365 258. Voloshyna I, Littlefield MJ, Kaplan L, Rieger JM, Figler R, Reiss AB (2013) Adenosine A2A receptor agonists regulate cholesterol homeostasis in mouse bone marrow derived macrophages (BMDM). FASEB J 27 259. Reiss AB, Grossfeld D, Kasselman LJ, Renna HA, Vernice NA, Drewes W, Konig J, Carsons SE, De Leon J (2019) Adenosine and the cardiovascular system. Am J Cardiovasc Drugs 19: 449–464 260. Gregory KJ, Bridges TM, Gogliotti RG, Stauffer SR, Noetzel MJ, Jones CK, Craig WL, Conn PJ, Niswender CM (2019) In vitro to in vivo translation of allosteric modulator concentrationeffect relationships: implications for drug discovery. ACS Pharmacol Transl Sci 2:442–452 261. Lane JR, May LT, Parton RG, Sexton PM, Christopoulos A (2017) A kinetic view of GPCR allostery and biased agonism. Nat Chem Biol 13:929–937 262. Deganutti G, Moro S (2017) Estimation of kinetic and thermodynamic ligand-binding parameters using computational strategies. Future Med Chem 9(5) 263. Zhang J, Yan W, Duan W, Wüthrich K, Cheng J (2020) Tumor immunotherapy using A2A adenosine receptor antagonists. Pharmaceuticals 13(9):237

46

D. Catarzi et al.

264. Augustin RC, Leone RD, Naing A, Fong L, Bao R, Luke JJ (2022) Next steps for clinical translation of adenosine pathway inhibition in cancer immunotherapy. J Immunother Cancer 10(2):e004089 265. Anighoro A, Bajorath J, Rastelli G (2014) Polypharmacology: challenges and opportunities in drug discovery. J Med Chem 57(19):7874–7887 266. Ma H, Huang B, Zhang Y (2020) Recent advances in multitarget-directed ligands targeting Gprotein-coupled receptors. Drug Discov Today 25(9):1682–1692 267. Zhou J, Jiang X, He S, Jiang H, Feng F, Liu W, Wei Q, Sun H (2019) Rational design of multitarget-directed ligands: strategies and emerging paradigms. J Med Chem 62:8881–8914 268. Morphy R, Rankovic Z (2005) Designed multiple ligands. An emerging drug discovery paradigm. J Med Chem 48(21):6523–6543 269. Wang M, Hou S, Wei Y, Li D, Lin J (2021) Discovery of novel dual adenosine A1/A2A receptor antagonists using deep learning, pharmacophore modeling and molecular docking. PLoS Comput Biol 17(3):e1008821 270. Barlow N, Baker SP, Scammells PJ (2013) Effect of linker length and composition on heterobivalent ligand-mediated receptor cross-talk between the A1 adenosine and beta2 adrenergic receptors. ChemMedChem 8(12):2036–2046 271. Pulido D, Casadó-Anguera V, Gómez-Autet M, Llopart N, Moreno E, Casajuana-Martin N, Ferré S, Pardo L, Casadó V, Royo M (2022) Heterobivalent ligand for the adenosine A2Adopamine D2 receptor heteromer. J Med Chem 65(1):616–632 272. Hagenow S, Affini A, Pioli EY, Hinz S, Zhao Y, Porras G, Namasivayam V, Müller CE, Lin J-S, Bezard E, Holger S (2021) Adenosine A2AR/A1R antagonists enabling additional H3R antagonism for the treatment of Parkinson’s disease. J Med Chem 64(12):8246–8262 273. Brunschweiger A, Koch P, Schlenk M, Rafehi M, Radjainia H, Küppers P, Hinz S, Pineda F, Wiese M, Hockemeyer J, Heer J, Denonne F, Müller CE (2016) 8-Substituted 1,3-dimethyltetrahydropyrazino [2,1-f]purinediones: water-soluble adenosine receptor antagonists and monoamine oxidase B inhibitors. Bioorg Med Chem 24(21):5462–5480 274. Ceni C, Catarzi D, Varano F, Dal Ben D, Marucci G, Buccioni M, Volpini R, Angeli A, Nocentini A, Gratteri P, Supuran CT, Colotta V (2020) Discovery of first-in-class multi-target adenosine A2A receptor antagonists-carbonic anhydrase IX and XII inhibitors. 8-Amino-6aryl-2-phenyl-1,2,4-triazolo [4,3-a]pyrazin-3-one derivatives as new potential antitumor agents. Eur J Med Chem 201:112478 275. Durante M, Squillace S, Lauro F, Giancotti LA, Coppi E, Cherchi F, Di Cesare ML, Ghelardini C, Kolar G, Wahlman C, Opejin A, Xiao C, Reitman ML, Tosh DK, Hawiger D, Jacobson KA, Salvemini D (2021) Adenosine A3 agonists reverse neuropathic pain via T cellmediated production of IL-10. J Clin Invest 131:e139299

Top Med Chem (2023) 41: 47–88 https://doi.org/10.1007/7355_2023_163 © The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 Published online: 19 August 2023

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year History of Pyrazolo[4,3-e][1,2,4]Triazolo [1,5-c]Pyrimidines (PTP) Filippo Prencipe, Tatiana Da Ros, Eleonora Cescon, Ilenia Grieco, Margherita Persico, Giampiero Spalluto, and Stephanie Federico Contents 1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.1 A1 AR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.2 A2A AR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.3 A2B AR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 1.4 A3 AR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 Pyrazolo-Triazolo-Pyrimidines (PTP) as AR Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.1 A2A AR Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.2 A3 AR Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2.3 A2B and A1 ARs Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3 Probes and Functional Conjugates Targeting Adenosine Receptors Bearing a PTP Scaffold . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1 Covalent Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2 Radiolabeled Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.3 Conjugated Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

48 49 50 50 51 53 55 59 65 67 67 69 72 77 78

Abstract Adenosine receptors are G protein-coupled receptors (GPCRs) involved in several processes in the human body. Their involvement in pathologies like coronary vasodilation, neurodegenerative diseases, inflammation, cancer, and pain make them interesting therapeutic targets. In particular, antagonism at the A2A and A3 adenosine receptor subtypes is of particular interest due to the central role in Parkinson’s disease and immunoncology of the first, and in cancer, glaucoma, and inflammation of the second. Pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidine nucleus is one of the most studied scaffolds to develop A2A and A3 adenosine receptor antagonists. In fact, a well distinct structure–activity relationship exists for these F. Prencipe, T. Da Ros, E. Cescon, I. Grieco, M. Persico, G. Spalluto, and S. Federico (✉) Department of Chemical and Pharmaceutical Sciences, University of Trieste, Trieste, Italy e-mail: [email protected]

48

F. Prencipe et al.

receptors. As a consequence, pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidine-based antagonists have also represented an optimal starting point to design probes and multifunction molecules that potently and selectively target A2A and A3 adenosine receptors. In this review, we reported the structure–activity relationship of pyrazolo [4,3-e][1,2,4]triazolo[1,5-c]pyrimidines at all the adenosine receptors along with all functionalization performed till now with their applications to study the structure and function of A3, but especially A2A adenosine receptors. Thus, pyrazolo[4,3-e][1,2,4] triazolo[1,5-c]pyrimidines and adenosine receptors are simply examples that would serve as a model that could be applied to every GPCR when a robust structure– activity relationship of a ligand class is known. Keywords Adenosine receptors, Binding, Carbon nanotubes, Dendrimers, Fluorescent ligands, GPCR, Preladenant, Probes, Pyrazolo[4,3-e][1,2,4]triazolo [1,5-c]pyrimidines, Radioligands, Radiotracers, SCH442416, SCH58261

1 Introduction Adenosine is an endogenous nucleoside widely distributed in mammalian organisms where it modulates several important physiological functions [1]. It derives from the degradation of intracellular adenosine monophosphate (AMP) and it is released from cells by a concentration-dependent, bi-directional nucleoside transporter, and it may then exert its extracellular effects. The adenosine produced from extracellular AMP by ecto-5′-nucleotidases may also contribute to rise extracellular adenosine levels [2]. Adenosine is removed from the extracellular space through the action of specialized transport proteins [3], while, intracellularly, it may be degraded by adenosine deaminase (ADA) to inosine, which is then transformed into uric acid. Adenosine possesses various properties similar to neurotransmitters, in particular: (1) it acts on specific receptors; (2) it can be antagonized by specific compounds; (3) enzymes producing adenosine are present in synapses; (4) its effect could be terminated by an efficient reuptake and a metabolic pathway. Nevertheless, adenosine is usually considered a neuromodulator because there is no evidence that it is stored in or released by specific purinergic nerves [4]. Adenosine’s role appears to be a homeostatic regulation under normal, physiological conditions, and a protective role in emergency situations [5]. Furthermore, adenosine could indirectly influence the action of neurotransmitters and other neuromodulators, behaving as a modulator of modulators. It could be considered as a fine-tuner and, in this way, it contributes to a very sophisticated interplay between its own receptors and the receptors for other neurotransmitters or neuromodulators [6]. Adenosine action occurs through the stimulation of the purinergic receptors that were classified as P1 receptors, while the receptors activated by phosphorylated nucleotides were classified as P2 receptors [7]. The P1 family could be subdivided into four receptor subtypes named A1, A2A, A2B, and A3 adenosine receptors (ARs),

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

49

respectively, which have been cloned and pharmacologically characterized. All these receptor subtypes are G protein-coupled receptors (GPCRs) and can be distinguished according to their preferred mechanism of signal transduction. A1 and A3 ARs interact with pertussis toxin-sensitive G proteins of the Gi and Go family, while the canonical signaling mechanism of A2A and A2B ARs is the stimulation of adenylyl cyclase via Gs proteins. The consequence of this activation is a reduction (A1 AR, A3 AR) or an increase (A2A AR, A2B AR) of the levels of cyclic adenosine monophosphate (cAMP) as second messenger. In addition to the coupling to adenylyl cyclase, all four subtypes may positively couple to phospholipase C via different G protein subunits [1, 8]. It has also been demonstrated that adenosine receptors could activate signal pathways that are not related to G proteins, in particular the β-arrestin one, inducing responses different from the G protein signals [8]. Considering the large distribution of adenosine in the organism, it is evident that adenosine receptors could be a significant target for several pathologies and in fact, in the last decades, different classes of potent and selective derivatives, both agonists and antagonists, have been synthesized with the aim to characterize and study the physio-pathological role of these receptors and their possible role in several disorders [9, 10].

1.1

A1 AR

Several studies have demonstrated the role of A1 ARs in respiratory diseases. As an example, the A1 AR antagonism blocks allergic responses to house dust mites in allergic rabbit model of asthma [11]. In contrast, in mice, A1 AR also mediates antiinflammatory effects via macrophages and reduces polymorphonuclear infiltration by inhibiting the release of chemotactic cytokines [12, 13]. Moreover, it has been demonstrated that A1 AR agonists decreased inflammation, edema, and neutrophil chemotaxis [14] and these receptors promoted the recruitment of leukocytes to infected lungs and attenuated their injury [15]. In inflammation processes, there are evidences that A1 AR activation could induce both proinflammatory and antiinflammatory responses [16]. Low concentration of adenosine induces neutrophil chemotaxis and adherence to endothelium via A1 AR [17], in contrast, A1 AR resulted to be a potent anti-inflammatory mediator in various kidney, heart, lung, and brain injury models [18]. In the cardiovascular system, A1 AR mediated negative chronotropic and dromotropic effects through inactivation of the inwardly rectifying K+ current, inhibition of the inward Ca2+ current, and activation of nitric oxide synthase. Furthermore, A1 AR activation showed both antiarrhythmic and proarrhythmic effects [19] and various A1 AR agonists have been evaluated as antiarrhythmic agents [20–23]. In the central nervous system (CNS), A1 ARs contribute to neuroprotection but they are also involved in neurodegeneration. In fact, their activation under hypoxia

50

F. Prencipe et al.

leads to the inhibition of presynaptic Ca2+ influx-related release of neurotransmitters, in particular glutamate, to generate neuroprotection [24, 25]. Nevertheless, it was assumed that prolonged activation of A1 AR increased A2A AR levels, which generate neurodegeneration in ischemic stroke [26]. It has also been observed that the use of A1 AR agonists amplifies nociceptive thresholds in spinal glial in neuropathic pain [27], it is also able to localize epileptic foci [28], and it decreases tremors in Parkinson’s disease [29]. Moreover, A1 ARs play a significant role in some metabolic diseases [30], in fact their agonists have potential use as antilipolytic agents, and, they are under development for the treatment of type 2 diabetes mellitus [31].

1.2

A2A AR

A2A ARs act as the most dominant anti-inflammatory effectors of extracellular adenosine [32], in fact the activation of these receptors inhibits neutrophil adhesion to endothelial cells, and formation of reactive oxygen species [16]. In the respiratory system, their activation affects multiple aspects of inflammatory process, in particular in asthmatic airways suppressing inflammation by reduced neutrophil adherence to the endothelium, and, moreover, there is the inhibition of histamine and tryptase release. In fact, in animal models of asthma and chronic obstructive pulmonary diseases (COPD), A2A AR agonists have resulted in good outcomes [33, 34]. In the cardiovascular system, A2A ARs seem to protect against ischemia and increase the occurrence of cardiac arrhythmias [35]. In the CNS, A2A ARs are in general associated to neurodegeneration, due to their excitatory effects [36]. Nevertheless several ligands, both agonists and antagonists, are under investigation for the treatment of several CNS disorders. In particular, agonists are under investigations for sleep disorders [36], schizophrenia [37], pain [38, 39], and drug addiction [40], while antagonists for the treatment of stroke [41], anxiety/depression [42], Alzheimer [43], Huntington [44], Parkinson [45–47], schizophrenia [37], and epilepsy [48]. At the present, the most appealing use of A2A AR antagonists is in cancer immunotherapy. In fact, it has been demonstrated that adenosine is capable to create a protection in the tumor microenvironment for both solid and hematologic tumors [49]. Adenosine acts both on A2A and A2B ARs inducing an anti-inflammatory phenotype in T cells, macrophages, and other cells, and antagonists have a beneficial effect when combined with immunotherapy [50].

1.3

A2B AR

A2B ARs are expressed on most inflammatory cells and possess both proinflammatory and anti-inflammatory effects [51]. In general, they generate antiinflammatory responses by coupling with Gs protein and proinflammatory effects by

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

51

coupling with Gq protein [52]. For these reasons, the use of agonists or antagonists for the treatment of inflammatory diseases is still controversial [53]. As mentioned, A2B AR binds both with Gs and Gq proteins to mediate airway reactivity, inflammation, and asthma [33]. Activation of A2B ARs in mast cells induces release of inflammatory mediators with bronchoconstriction as a consequence [54]. In fact, A2B AR antagonists prove to be effective in the treatment of asthma in mouse model [55]. A2B AR agonists reduce pulmonary edema and improve histologic lung injury, diminishing lung inflammation [56, 57]. The studies performed to clarify the role of A2B AR in the cardiovascular system are conflicting. It seems that A2B AR plays an important role in ischemia preconditioning [58] but, at the same time, other studies reported that A2B AR is not involved [59]. A2B ARs have also been proposed to inhibit smooth muscle cellassociated hypertension [60], and they may contribute to the pathogenesis of atherosclerosis by preventing atherosclerosis plaque formation [61]. It is also known that A2B ARs are involved in homeostasis and lipid metabolism, insulin secretion and resistance, β-cell survivor and kidney protection, and the use of agonists, in fact, inhibits diabetes and protects pancreas [62]. A2B ARs are also highly expressed in tumor cells and promote tumor cell proliferation; in fact, their antagonists seem to reduce cell growth in tumors and metastasis, permitting to consider A2B AR as a target for novel or combined therapies in cancer [63, 64].

1.4

A3 AR

The dual effect of A3 ARs in the inflammation process involves many cells that can have overlapping and opposite function [16]. A3 ARs produce a proinflammatory effect by promoting the release of histamine and other allergic mediators [65, 66]. Nevertheless, A3 ARs inhibit lipopolysaccharide (LPS)-stimulated release of tumor necrosis factor-α (TNF-α) and nitric oxide (NO), by increasing neutrophil chemotaxis and, in contrast, mediate the inhibition of oxidative burst in human neutrophil and promyelocytic HL60 cells [67, 68]. Clinical trials demonstrated that A3 AR allosteric modulators induce an antiinflammatory effect in animal models of arthritis [69], and the use of A3 AR agonists could be useful for treatment of patients with moderate to severe plaque psoriasis [70]. In contrast, the use of A3 AR antagonists inhibits shrinkage of human non-pigmented ciliary epithelial cells and reduces mouse intraocular pressure in an animal model of glaucoma [71, 72]. A3 ARs also play a role in asthma, chronic obstructive pulmonary disease (COPD), lung fibrosis, and pulmonary inflammation. Nebulized agonists directly induced lung mast cell degranulation, while having no effect in adenosine receptor knock-out (KO) mice [73]. Taking into account that A3 ARs are not present in human lung mast cells and that they inhibit only degranulation of eosinophils, they could be useful only in

52

F. Prencipe et al.

eosinophil-dependent allergic disorders. On these bases, we could assert that A3 ARs are involved in both proinflammatory and anti-inflammatory responses depending on the cell type involved [74]. Activation of the A3 ARs either prior to ischemia or during reperfusion is useful in cardioprotection: [75] administration of agonists before ischemia or during reperfusion reduces infarct size or inhibits myocardial ischemia-reperfusion injury [76, 77]. In contrast, other experimental observations clearly indicate that low-level expression in the heart provides effective protection against ischemic injury without adverse effects, while higher levels lead to the development of a dilated cardiomyopathy [78]. In the last years, it has been demonstrated that selective A3 AR antagonists could be useful for the treatment of atherosclerosis, and hypercholesterolemia, and could ameliorate diabetic kidney complications [79]. Nevertheless, the role of cardioprotection by A3 ARs is not yet completely clarified [80]. The expression of A3 ARs in the brain is low but several studies have reported a neuroprotective function [81], as in ischemia. In this case, it is possibly mediated by several pathways including microtubule-associated protein 2 (MAP-2), enhancement of the expression of glial fibrillary acidic protein, depression of nitric oxide synthase (NOS), stimulation of glial C-C motif chemokine ligand 2 (CCL2) synthesis, and delay of irreversible synaptic failure [82, 83]. Very recently an interesting application of A3 AR agonists for the treatment of neuropathic pain has also been reported [84]. In contrast, the use of antagonists seems to increase the stability of IGABA in different epileptic tissues, giving new opportunities in the treatment of epilepsy [85]. A3 ARs seem to be involved in the protection in inflammatory gastrointestinal diseases, in fact the treatment with agonists reduces oxidative damage and inflammatory mediators in the colon [86]. Instead, the use of A3 AR antagonists produces protection in renal failure [87]. A3 ARs play an important role in cancer, in fact primary and metastatic solid tumors show high expression of this receptor subtype [88], and it is well known that A3 ARs are involved in the regulation of the cell cycle. In the event of tumor inhibition, the antiproliferative effect occurs mainly via A3 ARs-induced cell cycle arrest in the G0/G1 phase and decreases in telomeric signaling in these cells. It has been demonstrated that agonists protect the retinal ganglion by increasing survival and they mediate a tonic proliferative effect in colon tumor cells [89–93]. A3 AR agonists inhibited tumor cell growth of HCT-116 human and CT-26 murine colon carcinoma cell and B16-F10 melanoma cells, and in in vivo studies have shown that in PC-3 prostate carcinoma cells and in N1S1 rat hepatocellular carcinoma cells induced apoptosis and tumor growth inhibition [94–96].

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

53

Fig. 1 5-Amino-9-chloro-2-(2-furyl)1,2,4-triazolo[1,5-c]quinazoline and pyrazolo[4,3-e][1,2,4] triazolo[1,5-c]pyrimidines

2 Pyrazolo-Triazolo-Pyrimidines (PTP) as AR Antagonists PTP tricyclic scaffold represents the pharmacophoric core of several series of non-xanthine heterocyclic compounds displaying antagonistic activity at A2A and A3 ARs. The initial studies aimed at the discovery of potent and selective adenosine receptor antagonists led to the identification of several classes of non-xanthine heterocyclic compounds possessing antagonistic properties either at A1 or A2A adenosine receptors such as triazoloquinazolines [97], triazoloquinoxalines [98], and imidazoquinoline [99]. Among these structurally diverse chemical entities, compound 5-amino-9-chloro-2-(2-furyl)1,2,4-triazolo[1,5-c]quinazoline derivative GCS 15943 (1, Fig. 1) was reported to exert high potency as A2A receptor antagonist while showing low selectivity at the A2A AR vs A1 AR [97]. On the basis of these findings and with the aim to enhance potency and A2A selectivity, in 1993, Gatta et al. explored new chemical entities reporting on the first series of pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidine derivatives as A2A adenosine receptor antagonists (PTP 2 a-b) (Fig. 1) [100]. The pyrazolo[4,3-e][1,2,4] triazolo[1,5-c]pyrimidine derivatives described in the paper exhibited nanomolar affinity at the A2A AR, representing the first step for the development of a new class of potent and selective AR antagonists.

54

F. Prencipe et al.

Fig. 2 Structures of AR antagonists as tools for functional characterization

Following this preliminary work, great efforts by medicinal chemists and pharmacologists have been made in order to produce more potent and selective A2A and A3 AR antagonists. Beginning in 1994, several series of PTP have been reported, some with good selectivity and potency [101–113]. Given the vastness of the subject, the synthetic approaches employed for the synthesis and the related optimization studies of pyrazolo-triazolo-pyrimidine derivatives are behind the scope of this review but a detailed dissertation can be found elsewhere [114]. Among the most advanced PTP derivatives, preladenant (Scheme 420814/MK3814, 3, Fig. 1), an A2A AR antagonist arising from an optimization study of the PTP tricyclic scaffold, has reached the phase III clinical trial for the treatment of Parkinson’s disease (PD), but failed for lack of efficacy (Fig. 1) [109, 115–117]. The discovery of potent and selective AR antagonists over the years has provided researchers important tools (Fig. 2) for the characterization of the physiological/ pathological role of the ARs and their distribution in mammalian organisms. One of such compounds is Scheme 58261 (4), a PTP derivative synthesized by Baraldi et al. [101, 102]. Scheme 58261 has been shown to have an affinity in the low nM range (Ki value of 1–2 nM) for A2A ARs expressed by a variety of tissues and cell types, and a good A2A vs A1 ARs selectivity (about 50- to 100-fold in rat and bovine brain, respectively), whereas it has no or little affinity for A2B and A3 ARs [118]. Its tritiated form has been used to specifically label A2A ARs, in particular regions of the rat brain such as caudate-putamen, nucleus accumbens, and tuberculum olfactorium [119]. Scheme 58261 (4) antagonizes in a competitive manner the effects of rabbit platelet aggregation and porcine coronary artery relaxation induced by the A2A AR selective agonist CGS 21680 2-[4-(2-carboxyethyl)-phenethyl-amino]-5′-Nethylcarboxamidoadenosine [118]. In rat, after peritoneal administration, Scheme 58261 (4) has demonstrated to counteract the effect of A2A AR agonists, enhancing locomotor activity [120], increasing waking behavior [121], and slightly increasing both blood pressure and heart rate [122]. In rat models mimicking CNS disorders, selective blockade of A2A AR by Scheme 58261 (4) potentiates the antiparkinsonian effects of L-dopa, indicating a therapeutic potential of this class of compounds in PD treatment [123, 124]. A2A AR antagonists have been considered attractive tools to improve the treatment of neurological disorders by preventing neurons damage and Scheme 442416 (5), described by Baraldi et al. [104], has been shown to increase the glutamate uptake activity in retinal Muller cells under increased hydrostatic pressure. This

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

55

molecule, deriving from the same family of PTP compounds, increased expression levels of glutamine synthase and glutamate aspartate transporter thus acting as neuroprotective agent, and it has been considered a potential candidate for the treatment of glaucoma by accelerating the clearance of extracellular glutamate [125]. Moreover, it displayed binding with different affinity to particular populations of postsynaptic A2A ARs in the rat striatum, suggesting the existence of at least two functionally and pharmacologically different populations of striatal postsynaptic A2A ARs [126]. Instead, the pharmacological characterization of the human A3 AR has been achieved by using MRE 3008F20 (6), a highly selective and high-affinity human A3 AR antagonist [105, 127]. In fact, it has been shown to display high selectivity for human A3 AR (Ki = 0.85 nM) compared with its affinity to human A1 (Ki = 1,100 nM), A2A (Ki = 140 nM), and A2B (Ki = 2,100 nM) ARs [128]. It has been employed to investigate the presence of the A3 AR on human colon cancer cells, and to evaluate its functional effect on colon cancer cell biology. The adenosine-mediated stimulatory effect, which results in proliferative effect, was reduced by MRE 3008F20, suggesting a potential role of the A3 AR in human colon cancer cell lines such as Caco2, DLDI, and HT29 (Fig. 2) [93].

2.1

A2A AR Antagonists

The first series of PTP derivatives as AR antagonists has been described by Gatta and coworkers [100]. As depicted in Fig. 3, in this preliminary study, the synthesis of five different PTP scaffolds has been described: 7-substituted-3,7-dihydro-2Hpyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-2-one (7), 2,7-disubstituted-7Hpyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidine (8), 5-amino-2,7-disubstituted-7Hpyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidine (9), and 5-amino-2,8-disubstituted8H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidine (10). Among the synthesized compounds, the most promising A2A AR antagonists were found in the 5-aminopyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidines series (9,10). Specifically, compound 8-FB-PTP (11) showed the highest affinity at the A2A AR (hA2A Ki = 1.2 nM), whereas 7-FB-PTP (12) displayed a slight decrease in affinity at the A2A AR (hA2A Ki = 12 nM) but a better selectivity against the A1 AR than compound 11 (12, A1/ A2A = 15.8 vs 11, A1/A2A = 2.8) [100, 102]. Both compounds bear a free amino group at the N5 position, a 4-fluorobenzyl moiety at the 7 or 8 positions, and a 2-furyl ring at the 2 position. Free amino and furyl groups at the N5 and 2 positions, respectively, were essential for high affinity at both A1 and A2A ARs (Fig. 3). Several substitutions on the pyrazole ring, at the 7 and 8 positions, with alkyl or aralkyl moieties were investigated indicating that substitution at the 8 position, in general, resulted in compounds with good affinity for adenosine receptors but with low potential in discriminating different AR subtypes. Substitution in 7 leads to improved selectivity at the A2A AR as demonstrated by comparing compound 4 with its corresponding 8-β-phenylethyl derivative, rA1/rA2A = 52.6 and 3.4, respectively.

56

F. Prencipe et al.

Fig. 3 PTP derivatives as A2A AR antagonists: general structures 7–10 and compounds 11, 12, 4, and 13–19

Moreover, among 7 substituted derivatives, compounds bearing an arylalkyl moiety (e.g., 7-(3-phenyl)propyl, compound 13, SCH63390) were found to be superior in terms of selectivity at the A2A AR than compounds bearing an alkyl moiety (e.g., 7-n-butyl, compound 14) [101, 102]. In an in vivo cardiovascular model, SCH58261 (4) retained antagonist activity at the A2A AR [102]. Compounds 11–14 and compound 4 are highly lipophilic, therefore exhibiting poor water solubility, which is a common drawback for A2A AR antagonists. So, modifications on the aryl group of the substituent in 7 have been investigated by introducing several polar moieties in order to improve the hydrophilic character of

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

57

PTP derivatives while retaining high affinity and selectivity at the hA2A AR, which seems related to the phenyl ring [104, 107]. Moreover, the propyl chain at the 7 position, used to introduce the aryl moiety, was confirmed as best length for the linker. The proposed compounds displayed high affinity for hA2A AR with different degrees of selectivity over the hA1 AR. On the contrary, all compounds were found to be inactive at the hA2B and hA3 ARs. Hence, the introduction of a hydroxyl function at the para position of the phenyl ring of Scheme 63390 (13) resulted in both higher affinity and selectivity at the rA2A AR (compound 15). In the same way, the para-methoxy analog, Scheme 442416 5, was found to be a potent and selective A2A AR antagonist, both at the human and rat receptors (rA2A Ki = 5.3 nM, rA1/ rA2A = 487; hA2A Ki = 2.7 nM, hA1/hA2A = 611) [104]. In addition, different substituents on the phenyl ring, like basic or acidic functions which allow the preparation of soluble salts or prodrugs, were evaluated. The para-amino (16) and the para-methanamino (17) derivatives showed high affinity and selectivity at the hA2A AR (compound 16, hA2A Ki = 0.22 nM, hA1/hA2A = 9,818; compound 17, hA2A Ki = 0.13 nM, hA1/hA2A = 4,430) but no improvements were obtained in terms of water solubility. The introduction of a sulfonic acid group (compound 18) produced a marked increase in water solubility but at the expense of both the affinity and the selectivity at the hA2A AR, which were significantly reduced (18, hA2A Ki = 140 nM, hA1/hA2A = 1). An attempt to retain the water solubility of sulfonic derivatives while restoring affinity and selectivity at the hA2A AR was made by introducing a weaker acidic function such as carboxylic acid. Compound 19 displayed an acceptable affinity and selectivity at the hA2A AR (19, hA2A Ki = 4.63 nM, hA1/hA2A = 1,064) but the water solubility was lower than compound 18 [107]. Unfortunately, the strategy of producing water-soluble compounds by introducing a polar group on the phenyl ring connected to the PTP scaffold through the alkyl chain proved to be unsuccessful; therefore, a different approach was explored. Modifications were directed to the 7-arylalkyl side chain where the distance between the alkyl chain at the 7 position and the phenyl ring was increased by introducing a polar piperazine ring as spacer, with the introduction of various substituents on the phenyl ring, especially at the para position. The arylpiperazine derivatives displayed both high A2A binding affinity in the low nanomolar range and high selectivity over A1 receptor (Fig. 4) [109]. Additionally, the majority of derivatives demonstrated considerable oral activity in the rat haloperidol-induced catalepsy model, being considered as potential therapeutic agents for PD. Unsubstituted phenylpiperazine 20 showed potent oral activity 1 h after dose administration but the activity was lost after 4 h, due to metabolic conversion to derivative 21, which resulted inactive when tested at an oral dose of 1 mg/kg even though its high affinity at the A2A AR (hA2AKi = 1.3 nM). The in vivo metabolism issue was tackled by substituting the phenyl ring with moieties, like fluorophenyl unit, that are not prone to metabolism. One of such compounds, the 2,4-difluoro analog (Scheme 412348, 22) displayed potent oral activity but its water solubility was poor. Compounds with improved solubility were prepared by introducing etherlinked substituents on the phenyl ring leading to derivatives endowed with

58

F. Prencipe et al.

Fig. 4 PTP derivatives as A2A AR antagonists: compounds 20–24

subnanomolar affinity at the hA2A AR and prolonged oral anti-cataleptic activity. The methoxy derivative Scheme 420814/MK-3814 (3), for example, exhibited high affinity and selectivity profile (hA2A Ki = 1.1 nM, rA2A Ki = 2.5 nM; hA1/ hA2A = 1,340), robust oral anti-cataleptic activity in the rat model, and good solubility in water as crystalline free base (0.2 μM at native pH of 5.1 and 2 mM in 0.01 N HCl). For these reasons, it was subjected to extensive profiling and additionally further evaluated in clinical trials under the name of preladenant [115–117]. Although several high-resolution crystal structures of the A2A AR were obtained, no structure in complex with preladenant has been reported, thus its exact binding mode and interactions within the receptor binding site are still unknown. Claff et al. have presented the first high-resolution crystal structure of A2A-PSB1bRIL, a thermostabilized A2A AR mutant harboring only a single point mutation, in complex with a fluorescent preladenant conjugate providing insights into the interaction of the preladenant scaffold with the orthosteric binding site of the receptor as discussed in the fluorescent conjugate section of this review (Fig. 12) [129]. In general, modifications regarding the 2 position of the PTP scaffold have been constrained by the observation that the presence of the furanyl group was a key requirement for an effective binding of the ARs and its replacement with phenyl or substituted phenyl or as long as with other heterocycles rings led to a marked loss of affinity at all the AR subtypes. In addition, in most cases within these series of PTP derivatives, substitution of the pyrazole ring took place at the 7 rather than at the 8 position and hence, the binding mode of these compounds might not be the same as for their N8 analogs [108]. Interestingly, in an additional effort to improve solubility with respect to preladenant (3), the pyrazolo[4,3-e]-1,2,4-triazolo[4,3-c]pyrimidin-3-one scaffold has been investigated, resulting in a novel class of potent and selective A2A AR antagonists (Fig. 4). Introduction of a vinyl group at the 7 position and furan replacement at the 2 position with a benzyl or 3-chlorobenzyl moieties (compounds

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

59

23 and 24, respectively) afforded potent and selective A2A AR antagonists (23, hA2A Ki = 0.3 nM, hA1/hA2A = 1,115; 24, hA2A Ki = 0.4 nM, hA1/hA2A = 356). However, these compounds were inactive in the rat catalepsy assay due to poor pharmacokinetic properties and exposure in rat brain compared to preladenant 3 [111].

2.2

A3 AR Antagonists

The A3 AR has been cloned from different species such as rat, human, sheep, mouse, and rabbit [130–133, 134], and significant differences in sequence homology for A3 receptors have been observed among species [131]. In particular, rat A3 AR is significantly different from human (73.8% of identical sequence) and behaves anomalously in ligand binding assays, where different affinities values were obtained at rat and human A3 AR, respectively, when comparing the same antagonist ligands [106, 137]. Moreover, tissue distribution of A3 AR is very different in rat as compared to man [135]. The starting point for the development of potent and selective A3 AR antagonists can be dated back in 1996 when Jacobson and coworkers reported on a series of triazoloquinazoline derivatives among which compound 1, GCS 15493, displayed a certain affinity for the human A3 AR (hA3 Ki = 14 nM). Acylation of the free amino group at position 5 of GCS 15493 (1) with a phenylacetyl group led to MRS 1220 (25), a potent but not highly selective A3 AR antagonist (rA3 Ki = 0.65 nM, A1/ A3 = 469, A2A/A3 = 80) [136]. In the same year, Baraldi and coworkers described a series of N6-(substituted-phenylcarbamoyl)adenosine-5′-uronamide derivatives, such as compound 26, acting as potent A3 AR agonists (Fig. 5) [137]. The pivotal step was the idea to functionalize the amino group at the 5 position of the PTP scaffold, known to confer antagonism activity at the A2A ARs, with the arylcarbamoyl moieties of potent derivatives within the adenosine-5′-uronamide A3 agonists series [105]. Interestingly the researchers described the exclusive preparation of the 8-substituted derivatives, since the coupling reaction with isocyanate, leading to the formation of the carbamoyl moiety, failed in the case of the 7-substituted PTP derivatives. The synthesized compounds displayed high affinity at the hA3 AR with the level of selectivity being influenced by the presence of both arylureido and (ar)alkyl substituents at the 5 and 8 positions, respectively (Fig. 6). This behavior is in contrast if compared with the corresponding N5-unsubstituted derivatives which were also assayed at the various AR subtypes, showing high affinity for the A2A AR as long with low selectivity vs the A1 AR and poor activity at hA3 AR [105, 138]. Specifically, the aforementioned difference could be clearly seen by comparing N5-unsubstituted derivative 27, which was a poorly selective A2A AR antagonist (hA2A Ki = 0.34 nM, A1/A2A = 2.9), with compound 28 where the amino group functionalization with the para-methoxycarbamoyl moiety afforded a potent and selective A3 AR antagonist (hA3 Ki = 0.20 nM, A1/A3 = 5,485, A2A/ A3 = 6,950) [138]. The following optimization efforts were focused on

60

F. Prencipe et al.

Fig. 5 PTP derivatives as A3 AR antagonists: compounds 25–33

modifications of the chain in 8 while maintaining either the 4-methoxyphenylcarbamoyl or the 3-chlorophenylcarbamoyl moieties at the N5 position. Several alkyl and arylalkyl moieties were evaluated in order to identify the best substitution pattern in terms of steric and lipophilic features for the specific recognition of the A3 AR subtype. From the obtained binding results it was quite evident that at the 8 position in the pyrazole ring the presence of a small alkyl chain,

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

61

Fig. 6 PTP derivatives as A3 AR antagonists: compounds 34–42, 44–45, and compound 43 as A2B AR antagonists

62

F. Prencipe et al.

such as methyl, endowed the newly synthesized PTP derivatives with high affinity at the A3 AR and good selectivity versus the other AR subtypes (i.e., compound 29, hA3AR Ki = 0.20 nM; A1/A3 = 5,485, A2A/A3 = 6,950) [138]. Next, a further investigation on the effect of diverse substituents (e.g., chloro, fluoro, bromo, methyl, nitro, sulfonic acid, trifluoromethyl) at the various positions on the phenyl ring of the N5-phenylcarbamoyl group combined with the presence of a small alkyl group at N8 position has been carried out. Interestingly, the synthesized compounds displayed affinity values at the A3 AR in the subnanomolar range (0.16–3.7 nM). In particular, the lower affinity value obtained for the para-sulfonic acid analog (compound 30, hA3 Ki = 25 nM) was explained by the presence of a hydrophobic environment in the A3 AR region surrounding the para position. Moreover, molecular modeling studies highlighted the presence of a steric control at the para and meta position which explained why bulky and polar substituents were not well tolerated at these positions [139]. Thus, the most potent and selective compound of the series was the unsubstituted derivative 31 displaying an A3 Ki value of 0.16 nM, being more than 2000-fold selective for A3 over A1 and A2A ARs. When the phenylureido moiety was replaced with a phenylacetyl moiety, a typical feature of MRS 1220 (25), the affinity at the hA3 AR was maintained, whereas a slight drop in selectivity was noted (compound 31 vs compound 32, Fig. 5). On the contrary, the shortening of the amide chain by deletion of a methylene group (compound 33) produced a drop of affinity at the A3 AR, while retention of selectivity was observed (33, hA3 Ki = 15.7 nM; hA1/hA3 = 129, hA2A/hA3 = 56) [140]. Although good results have been achieved in terms of potency and selectivity at the A3 AR, the major problem with PTP derivatives was still the typical low water solubility which hampered their use as pharmacological and diagnostic tools. Based on the molecular modeling study suggesting the presence of steric control around the para position, an attempt to improve water solubility was made by bioisosteric replacement of the phenyl ring of the phenylureido moiety at the N5 position with a pyridine ring, which in turn allowed the preparation of pyridinium hydrochloride salts. Moreover, the position and the basicity of the pyridine nitrogen, as long as the replacement of pyridine moiety with different heteroaryl groups, were evaluated (Fig. 6). The best substituent profile was found to be the 4-pyridinium hydrochloride salt, as highlighted by compound 34 which, when compared with the unsubstituted phenyl derivative 31, displayed an increased water solubility (31 Rm = 4.06 vs 34 Rm = 1.66, where Rm = log(1/Rf - 1); maximum concentration in water of compound 34: 15 mM) as well as enhanced potency and selectivity at the hA3 AR (34, hA3 Ki = 0.014 nM; hA1/hA3 = 25,357, hA2A/hA3 = 7,857) [141, 142]. To evaluate the pharmacological potential of this series of PTP derivatives, aqueous formulations at physiological pH, suitable for intravenous administration, were prepared. Despite the increased water solubility with respect to that of compound 31, at physiological pH the solubility of 34 dropped to less than 0.1 mg/ml. Thus, a new series of derivatives, in which the pyridine-4-yl moiety was replaced by a 1-(substituted)-piperidin-4-yl ring, was designed with the idea of evaluating the higher basicity of piperidine and its potential to form stable, water-soluble salts. The best binding profile was shown by compound 35 (hA3 Ki = 9.7 nM; A1/A3 = 351,

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

63

A2A/A3 > 515, Fig. 6) which remarkably exhibited a solubility of 8 mg/ml at physiological pH, allowing the preparation of stable aqueous solutions suitable for intravenous administration [112]. A more recent study reported on an in-depth evaluation of several polyfunctionalized chains such as alkylamino, arylalkylamino, heterocycles moieties replacing the aryl carbamoyl or arylacetyl moieties at the N5 position, providing useful information about the extracellular environment surrounding the N5 position of the PTP nucleus. These specific modifications proved to be successful as demonstrated by the newly synthesized compounds displaying affinity for the hA3 AR in the nanomolar and subnanomolar range with different degrees of selectivity over the other AR subtypes. The introduction at the 5 position of small branched alkylamines and several benzylamines produced good results in terms of affinity but at the expenses of selectivity over the other AR subtypes which was poor (e.g., compound 36, hA3 Ki = 131 nM; A1/A3 = 25, A2A/A3 = 47.4). The best results in terms of potency and selectivity were achieved through the synthesis of compound 37 bearing an (S) α-phenylethyl chain at the N5 position (37, hA3 Ki = 0.3 nM; A1/A3 = 1,127, A2A/A3 = 184) [143]. Analogously, Federico et al. explored a related modification at the 5 position by introducing several aminoesters (e.g., compound 38) obtaining novel PTP derivatives endowed with good affinity at the hA3 AR. Furthermore, the aminoester moiety represents an optimal linker allowing further functionalization of the carboxylic group in order to obtain more complex conjugated derivatives for diagnostic/theranostic purposes [144]. Although compound 38 was not as potent at the hA3 AR as previously reported antagonists, it was chosen for molecular modeling simulations to evaluate the possible interactions of these PTP derivatives within the hA3 AR binding site. As highlighted by the docking pose of compound 38 at the hA3 AR, which is the pose observed at the receptor binding site among four different binding modes isolated from the docking simulations, and the related energy fingerprint (Fig. 7), the ligand formed hydrogen bonds (HBs) with residue N250, established electrostatic interactions with N250 and L90 and several hydrophobic contacts especially with F168, L246, and I268. Van der Waals interaction with F168 is repulsive but the hydrophobic terms tended to overcome it. These data indicated that this particular binding mode could represent a plausible solution for the binding of these PTP derivatives at the hA3 AR binding site [144]. Taking into account the modifications at the 2 position, the indication that N8 substituted PTP derivatives might display a different binding mode compared to N7 substituted derivatives (showing high selectivity at the A2A AR), and considering that several tricyclic hA3 AR antagonists are functionalized with a phenyl ring at the position equivalent to that of furan in PTP derivatives [145], Cheong and coworkers proposed a new structure–activity relationship evaluation by designing and synthesizing a series of PTP derivatives bearing a (para-substituted)-phenyl moiety at the 2 position maintaining either methyl or phenyl-ethyl groups at 8 position and a free amino, phenylacetamide or (bis-)benzamide at the 5 position. The binding assay results clearly indicated that the free amino group at 5 position poorly discriminates among the AR subtypes (compound 39). On the contrary, when a phenylacetamido group was introduced at the 5 position, the resulting derivatives retained good

64

F. Prencipe et al.

Fig. 7 Per-residue energy histograms (panel a) and docking pose (panel b) of compound 38 at the hA3 AR. Reproduced from [144] with permission from the Royal Society of Chemistry

affinity towards the hA3 AR subtype and a notable increase in hA3 AR selectivity over other AR subtypes was observed in comparison to the 2-furyl PTP derivatives (e.g., compound 40, hA3 Ki = 0.241 nM; A1/A3 > 124,000, A2A/A3 = 415,000 vs compound 32, hA3 Ki = 0.81 nM, A1/A3 = 877, A2A/A3 = 522 or vs compound 31, hA3 Ki = 0.16 nM; A1/A3 = 3,713, A2A/A3 = 2,381 [110, 144]. Moreover, Okamura et al. prepared different fused 1,2,4-triazolo[1,5-c]pyrimidine derivatives, including 5-n-butyl-pyrazolo[4,3-e]-1,2,4-triazolo[1,5-c]pyrimidines (Fig. 6), confirming that both the type and the position of the substituent on the phenyl ring at the 2 position affected the potency and the selectivity at the hA3 AR over the other AR subtypes. A higher hA3 AR selectivity over the hA2A AR was observed when the phenyl ring was substituted at the para position rather than at the meta or ortho positions as well as in comparison with unsubstituted phenyl derivatives (41 (4-F-Ph), hA3 IC50 = 1.9 nM; A1/A3 = 321, A2A/A3 = 5,263 vs compound 42 (Ph), hA3 IC50 = 2.1 nM; A1/A3 = 12.9, A2A/A3 = 90.5) [72].

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

65

Interestingly, in a further study aimed at the identification of new non-xanthine hA2B AR antagonists, Federico et al. reported on a novel series of PTP derivatives in which the furyl group at the 2 position was replaced with a 1-(3-trifluoromethylbenzyl)1H-pyrazol-4-yl moiety, a substituent known to confer high affinity for the hA2B AR when introduced at the 8 position of xanthine derivatives such as CVT-6975 (43), a potent hA2B AR antagonist. Surprisingly, the synthesized hybrids were inactive at the A2B AR while behaving as potent and selective hA3 AR antagonists. Among the synthesized derivatives, the best results were obtained with compound 44, which was not as potent at the hA3 AR as its corresponding 2-furyl derivative 32, but demonstrated a higher selectivity at the hA3 AR over the other receptor subtypes, thus indicating that replacement of the furyl group at the 2 PTP position with longer chain and more sterically hindering group such as 1-(3-trifluoromethyl-benzyl)1H-pyrazol-4-yl moiety is also well tolerated, opening new possibilities in the development of even more potent and selective PTP derivatives as A3 AR antagonists [113]. Finally, Baraldi et al. investigated also the 9 position of the PTP scaffold by evaluating the effects produced upon the introduction of several substituents with different steric and hydrophilic/lipophilic properties, such as alkylamino, arylamino, alkylthio, or N-methylpiperazino moieties while maintaining the key features for antagonism, that are the methyl group at the 8 position and a free amino or phenylureido as long with arylacetamido moieties at the 5 position of the PTP scaffold for A2A AR antagonism and A3 AR antagonism, respectively. In general, the introduction of a substituent at the 9 position resulted in a complete loss of selectivity even though the receptor affinity was maintained. Among the different substituents screened, the methylthio group at the 9 position was the best tolerated, as for compound 45, which displayed nanomolar affinity at the four AR subtypes (45, hA1 Ki = 8.4 nM; hA2A Ki = 1.2 nM; hA2B Ki = 10.3 nM; hA3 Ki = 35 nM) [108].

2.3

A2B and A1 ARs Antagonists

Whilst the identification of several potent and selective antagonists has allowed to outline a clear SAR profile for the PTP related compounds at the A2A and A3 AR subtypes, the same level of understanding regarding the structural requirements has not yet been achieved to obtain highly potent and selective PTP ligands for of the A2B and A1 AR subtypes. Many efforts have been spent and some typical features could be identified among some potent, but unselective, PTP A2B antagonists reported so far. In general, substitution at the 8 position, rather than at the 7 position of the pyrazole ring, seems to be favorable for the interaction with the A2B AR (Fig. 8). Specifically, Baraldi et al. reported that N5 unsubstituted PTP derivatives with long alkyl chain or a phenylethyl moiety at the 8 position displayed interesting affinity at the A2B AR in the nanomolar range but with low selectivity over the other

66

F. Prencipe et al.

Fig. 8 PTP derivatives as unselective AR antagonists: compounds 45–49.a Displacement of specific [3H]-DPCPX binding at human A2B AR expressed in HEK-293 cells; b Displacement of specific [3H]-ZM241385 binding at human A2B AR expressed in chinese hamster ovary (CHO cells)

AR subtypes (e.g., compound 27, hA2B Ki = 5.1 nM; compound 46, hA2B Ki = 9.1 nM) [138]. The introduction of a γ-aminobutyramido at the 5 position (compound 47) resulted in a slight decrease of affinity at the A2B AR, while an enhanced selectivity was observed over the A2A AR subtype [146]. Further optimization studies of the substituent at the N5 position led to the observation that a combination of branched or arylalkyl chain at the 8 position together with arylacetyl moieties at the N5 position promotes the binding with the A2B AR. In particular, the presence of a bulky α-naphthylacetyl moiety at the N5 position, such as in compound 48, produced a significant increase in selectivity at the A2B AR over the other receptor subtypes resulting in the first potent and selective PTP A2B AR antagonist [147]. Nevertheless, further evaluation of the binding properties of compound 48 produced controversial results. In fact, in a subsequent study, the potency of several A2A AR antagonists was determined through displacement of specific binding of [3H]-ZM241385, a dual A2A/A2B antagonist, at human A2B ARs expressed in CHO cell. Surprisingly, in this specific assay, compound 48 displayed a Ki value at the hA2B receptor of 2,190 nM, which is in evident contrast with the Ki value of 20 nM previously reported. These results have casted doubts about the effectiveness of compound 48 as an A2B AR antagonist [148].

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

67

As discussed in the previous section, a hybridization approach has been employed to identify new non-xanthine hA2B AR antagonists by introducing at the 2 position of the PTP scaffold a 1-(3-trifluoromethyl-benzyl)1H-pyrazol-4-yl moiety, which, when present at the 8 position of xanthine derivatives such as CVT-6975 (43), confers high affinity for the hA2B AR. Unfortunately, this approach proved to be unsuccessful, being the newly synthesized compound inactive at the hA2B AR, but surprisingly this modification led to some potent and selective hA3 antagonists, such as compound 44 [113]. A similar behavior could be observed for the A1 AR too. In general, 8-substituted PTP analogs displayed a better binding affinity at this receptor subtype compared to the 7-substituted PTP derivatives while showing no selectivity at the A2B AR over the A2A AR in particular, and generally over the other AR subtypes. This could be clearly seen by comparing compounds 12 and 4 with compounds 11 and 27, which bear a 4-fluoro benzyl and a phenylethyl moiety at the 7 and 8 position, respectively (12, rA1 Ki = 189 nM; rA1/rA2A = 15.8 and 4, hA1 Ki = 549 nM; hA1/hA2A = 499 vs 11, rA1 Ki = 3.3 nM; rA1/rA2A = 2.8 and 27, hA1 Ki = 1 nM; hA1/hA2A = 3.22) [102, 105, 137]. Moreover, the introduction of a substituent at the amino group at the 5 position resulted in a reduced affinity at the A1 AR. In particular, compound 49 displayed an affinity about 120-fold lower compared to its N5 unsubstituted analog, compound 27 (49, hA1 Ki = 120 nM vs 27, hA1 Ki = 1 nM) (Fig. 8) [138, 147].

3 Probes and Functional Conjugates Targeting Adenosine Receptors Bearing a PTP Scaffold 3.1

Covalent Derivatives

Irreversible ligands represent an useful tool to study receptor functions and structure, such as physiological functioning, receptor reserve determination, and ligand binding site mapping [149]. In addition, covalent ligands are effective at stabilizing GPCRs to obtain X-ray crystal structures [150, 151]. At the A2A AR, reaction of SCH58261 (4) with fluorosulfonic acid affords the A2A AR irreversible antagonist 5-amino-7-[2-(4-fluorosulfonyl)phenylethyl]-2-(2-furyl)-pryazolo [4,3-e]-1,2,4-triazolo[1,5-c]pyrimidine (FSPTP, 50, Fig. 9), which has been used in coronary conductance experiments on guinea pig hearts, showing that there is a large receptor reserve, which justifies the high agonist-mediated coronary vasodilation, that evidently is not consistent with agonist affinity values [152]. Another useful method to study protein targets is photoaffinity labeling (PAL). Regarding GPCR, a photoaffinity probe is useful both to localize receptor and to have information about receptor binding sites [153]. In particular, identification of the crosslink site could be obtained after UV-mediated reaction with a solubilized protein followed by digestion and LC-MS/MS analysis. Compounds 51 and 52 (Fig. 9) were developed by replacing the fluorine atom in 8-FB-PTP (11) and 7-FB-PTP (12) with

68

F. Prencipe et al.

Fig. 9 PTP irreversible compounds

the photophore trifluoromethylphenyl diazirine (TPD) group. Fluorescent polarization binding assay has been performed on solubilized hA2A AR showing Ki values of 39.7 and 1,520 nM for compounds 51 and 52, respectively. Thus, PAL of the receptor was performed using compound 52 and Tyr2717.36 has been identified as the most likely cross-linked amino acid [154]. A PTP scaffold has also been used to develop irreversible antagonists for the A3 AR [106]. In particular, the design of covalent derivatives was based on the selective A3 AR antagonist, which bear a short linear alkyl chain at the N8 position combined with a 4-methoxyphenylureido moiety at the 5 position. The replacement of the methoxy group at the para position of the arylurea was proposed introducing two different electrophile groups: fluorosulfonyl and bis-(β-chloroethyl)amino groups. The first can react with any kind of nucleophiles, thus with both amines, alcohols or thiols, while nitrogen mustard is less reactive than fluorosulfonyl moiety and, consequently, it reacts preferably with amines. Among the series, only fluorosulfonyl compounds exhibited an irreversible mechanism of action towards A3 AR, with the best compound bearing a propyl moiety at the N8 position (53, Fig. 9). A3 ARs were preincubated with the compounds, washed to remove the free antagonist and then submitted to classical radioligand binding. Docking studies suggested that in the proximity of the transmembrane (TM) binding cleft there are at least two residues possibly implicated in the irreversible binding: Ser247 and Cys251. Even remaining more potent at the A3 AR (47% of inhibition at 1 nM and 70% of inhibition at 10 nM) compound 53 exhibited also a good affinity against A2A AR (Ki A2A AR = 50 nM) [106].

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

69

Fig. 10 PTP radioligands and radiotracers for the A2A AR (54–62) and A3AR (63)

3.2

Radiolabeled Derivatives

PTP derivatives have been used to develop radiolabeled compounds principally targeting the A2A AR subtype. Different radioactive atoms have been inserted as 3 H (tritium), 11C, 14C, and 18F (Fig. 10). Radioligands are developed both to study target receptor and for drug development, both in vitro and in vivo. Radioligands are

70

F. Prencipe et al.

used in various stages of drug development: simple screening by in vitro binding assays; demonstration of target engagement, distribution at the target tissue/organ; and evaluation of the relationship between plasma concentration and target occupancy in vivo. Last points are particularly important to determine the dose range in the clinic. In vivo studies were feasible thanks to positron emission tomography (PET) molecular imaging [155, 156]. Given the versatility of this approach, it is therefore obvious that a variety of probes containing radioactive atoms with different half-lives are required, depending on the specific application. The first developed tritium PTP has been [3H]Scheme 58261 (54). Due to its A2A AR selectivity observed in binding assays, this compound becomes a good radioligand for autoradiographic studies on receptors in rat, mouse, and human brains [119, 157–161]. In addition, it has been successfully applied in scintillation proximity assay (SPA)-based ligand screening [162]. [3H]Scheme 58261 (54) was found to label A2A receptors with high affinity (hA2A KD = 2.3 nM) and with a rapid, saturable, and reversible specific binding of 70% [158]. Another PTP A2A AR antagonist explored to obtain radioligands useful for PET imaging is Scheme 442416 (5). In this case, it was labeled with a 11C atom, [11C]Scheme 442416 (55). Because A2A AR antagonists are studied for the treatment of Parkinson’s disease, a radioligand for this receptor should be able to permeate blood-brain barrier along with high affinity and selectivity, low nonspecific binding, and minimal metabolism. Another key factor is that half-life of these PET radiotracers should be minimal, thus radiosynthesis needs to be easy and fast. [11C]Scheme 442416 (55) is easily obtained by direct alkylation of hydroxy derivative with [11C]CH3I under alkaline conditions. Reaction and purification require a total of 40 min with a radiochemical yield of 29% [163]. Studies on rats and primates identify [11C] Scheme 442416 (55) as a promising tracer for the in vivo PET imaging of A2A AR [164]. Because A2A AR density has been reportedly increased in the caudate–putamen in PD patients, A2A AR-mediated PET imaging can help for early identification of this disease [165]. With respect to 11C, 18F isotope has higher specific activity and a longer physical half-life (109.8 vs 20.4 min), thus providing longer biodistribution and scanning times, and, not least, it allows a distribution of the radiotracer to image centers without cyclotron facilities [166]. For these reasons, Bhattacharjee and coworkers developed a 18F-containing analog of Scheme 442416 (5), [18F] MRS5425 (56) where the methoxy group at para position of the N7-phenyl propyl moiety has been replaced by the 2-[18F]fluoroethoxy group (Fig. 10). [18F]MRS5425 ([18F]FESCH, 56) radioactivity in rat striatum, after reaching the maximum, decreases slower than with [11C]Scheme 442416 (55), demonstrating to be a good probe [167]. Also a 3-[18F]fluoropropoxy derivative, [18F]FPSCH (57), has been developed in order to evaluate the effect of a longer fluoroalkyl chain, but the authors concluded that these compounds performed equally well in rats [166]. In a study aimed to highlight changes in the availability of A2A/D2 heterodimeric receptors in a mouse model of Parkinson’s disease, [18F]MRS5425 (56) led to inconclusive data, at least in part due to the brain penetration of a non-negligible fraction of a single radiometabolite [168]. To overcome this problem, a deuterated isotopologue of [18F]

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

71

MRS5425 (56), [18F]FLUDA (58), with enhanced metabolic stability has been developed. As expected, no brain-penetrant radiometabolites have been detected [169, 170]. Obviously, after its discovery, preladenant (3) became the focus of the research on A2A AR antagonists for the treatment of PD. In particular, Merck, the proprietary of this molecule following its acquisition of Schering-Plough Corp, developed three isotopically labeled forms of this molecule. [3H]MK3814 (59) and [14C]MK3814 (60) were used for a preliminary and more definitive (e.g., in human) absorption, distribution, metabolism, and excretion data (ADME) evaluation of the compound, respectively. In this work also the non-radioactive deuterated compound [2H]MK 3814 (e.g., fully deuterated ethyl chain between pyrazole and piperazine) was prepared as an internal standard to be used in a liquid chromatography mass spectrometry-based bioanalytical method [171]. Preladenant (3) represents also a starting point to develop other new radiotracers. In particular, in order to obtain a single photon emission computed tomography (SPECT) radiotracer, a longer-lived isotope, 123I, has been used in analog [123I]MNI-420 (61). In fact, a 123I-based radiotracer could be dispensed from a central pharmacy rather than generated on-site [172]. During evaluation on humans, [123I]MNI-420 (61) demonstrated to possess the same characteristics than currently used SPECT radiotracers for other human neuroreceptors imaging (Fig. 10) [173]. In 2014, Barret et al. reported [18F] MNI-444 (62), obtained by reaction between the tosyl derivative 2-(4-(4-(2-(5-amino-2-(furan-2-yl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c] pyrimidin-7-yl)ethyl)piperazin-1-yl)phenoxy)ethyl-4-methylbenzenesulfonate and 18 F . [18F]MNI-444 (62) has been developed to investigate receptor occupancy and plasma levels of preladenant (3) and tozadenant, a benzothiazole-based A2A AR antagonists, in nonhuman primates (NHP) [174]. In these studies, compound 62 showed a better specific-to-nonspecific ratio than other A2A AR PET radiotracers available at that time, e.g. [11C]TMSX and [11C]Scheme 442416 (55), successively confirmed also in humans brain [174, 175]. Concerning A1 and A2B ARs both agonists and antagonists radioligands are reported in literature, but none bearing a PTP scaffold [176]. Instead, on A3 AR, MRE 3008F20 (6) has been successfully developed as tritiated derivative [3H]MRE 3008F20 (63) (Fig. 10) [128, 177]. [3H]MRE 3008F20 (63) has been used to detect the presence of A3 AR on plasma membrane of human lymphocytes, correlating the up-regulation of A3 receptors functionally coupled with adenylyl cyclase in activated T cells with a potential biological role for adenosine-mediated responses in the immune system [178]. Gessi et al. used this radioligand also to investigate A3 AR density in colon carcinomas, finding that it was higher than in normal mucosa of the same patient. Interestingly, overexpression of A3 AR in tumors was reflected in peripheral blood cells, indicating that this receptor could be a diagnostic marker for colon cancer [179].

72

3.3 3.3.1

F. Prencipe et al.

Conjugated Derivatives Fluorescent Derivatives

Fluorescent ligands are of great interest because they allow real-time monitoring of the localization and function of the receptors in unmodified human cells, both in health and disease, at endogenous expression levels and this knowledge is fundamental to develop new therapies [180]. In the last 10 years, PTPs were conjugated to different fluorophores obtaining fluorescent antagonists towards A2A and/or A3 ARs. Applications of these compounds range from binding assay to fluorescence microscopy and receptor detection. Taking inspiration from Scheme 442416 (5), Jacobson and coworkers designed the Alexa Fluor 488 derivative MRS5346 (64), which showed a Ki of 111 nM on A2A AR (Fig. 11). The problem in developing fluorescent ligands is that fluorophore conjugation should not interfere with the ligand recognition in the orthosteric binding site of the receptor. For compound 64, docking studies suggested that the linker and the fluorophore protruded towards the extracellular region of the A2A AR. In addition, MRS5346 (64) was successfully employed as ligand in a fluorescent polarization (FP) binding assay which represents an alternative to the classical radioligand-based binding assay [181]. Alexa Fluor 488 has a lifetime appropriated to be used in FP (4.5 ns); shorter lifetimes like in TAMRA (65) and BODIPY650/ 665 derivatives (MRS5418, 66) were not suitable for this application, but these fluorescent ligands could be used for other purposes like fluorescence microscopy [182]. The same group reported also other two Scheme 442416-fluorescent analogs, compounds 67–68, MRS7416 (69), and MRS7396 (70) bearing Alexa Fluor 647, Alexa Fluor 488, and BODIPY630/650-X as fluorophore moiety, respectively (Fig. 11). MRS7416 (69) and MRS7396 (70) display highest affinity at the A2A AR, thus has been subjected to docking studies using the high-resolution A2A AR X-ray structure (PDB ID: 4EIY). Molecular modeling analysis revealed that these compounds behave as bitopic ligands, where PTP resides in the orthosteric binding site of A2A AR, while fluorophores allocate in different adjacent clefts [183]. Subsequent work demonstrated that MRS7416 (69) and MRS7396 (70) bitopic ligands are allosteric antagonists; authors studied several different fluorescent ligands, demonstrating that they can behave both as allosteric or competitive antagonists, depending on the nature of the linker and fluorophore [184]. An approach similar to that proposed by Jacobson’s laboratory has been used by Kellam and coworkers on preladenant (3), where the binding pose at the hA2A AR revealed that the chain on the phenyl ring is oriented towards the more solventexposed part of the receptor binding cleft, thus representing a good anchoring point for a fluorophore moiety. These data have been confirmed obtaining different A2A AR fluorescent probes (71–76), based on the preladenant structure that maintained a good affinity towards the receptor (Fig. 11) [180]. Compounds 71–76 were applied in a bioluminescence resonance energy transfer (BRET) binding ligand assay,

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

Fig. 11 PTP fluorescent A2A AR antagonists

73

74

F. Prencipe et al.

Fig. 12 (a) Structure of the A2A AR fluorescent ligand PSB-2115 (77); (b) Ligand binding pocket of A2A-PSB1-bRIL-PSB-2115 (77) (PDB:7PYR) taken from Angew. Chem. Int. Ed. 2022, 61, e202115545 International Edition: doi.org/10.1002/anie.202115545 [129]. The 2Fo-Fc electron density of PSB-2115 is shown in orange mesh (contoured at 1.0 σ)

exhibiting a clear saturable component of specific binding along with low levels of nonspecific binding. Because one possible application of fluorescent ligands is in receptor visualization, compounds 71 and 74 were evaluated for this purpose in confocal microscopy. Images indicated a successful specific binding to A2A AR at the cell membranes with undetectable uptake into the cytosol. Finally, due to its optimal characteristics, which comprise a good water solubility, Alexa Fluor 647 derivative 76 has been further validated as fluorescent ligand in BRET binding assay of known hA2A AR antagonist, resulting in a promising pharmacological tool [180]. Recently, an X-ray crystallographic structure of the new preladenant-derived fluorescent A2A AR antagonist PSB-2115 (BODIPY as fluorophore, 77) has been reported by Claff et al. Unfortunately, no electron density could be observed for the linker and the fluorophore, but the pose of PTP scaffold in the binding pocket is identical to that reported for the precursor, demonstrating that BODIPY does not interfere with PTP binding to A2A AR [129]. As shown in Fig. 12, in this structure, preladenant portion of PSB-2115 (77) shows direct interactions to helices V, VI, VII and to the second extracellular loop (ECL2) of A2A AR. The typical key hydrogen bond networks between 5-amino group and N2536.55/E169ECL2 and between furan oxygen and N2536.55 can be observed, along with the tricyclic π–π stacking interaction with F168ECL2. Other important interactions are the hydrophobic contacts with L2496.51 and I2747.39. As predicted by previous computational studies, the phenylpiperazinylethyl moiety attached to the N7 in the PTP scaffold effectively extends towards the extracellular surface of the A2A AR, stabilized by π–π stacking to H264ECL3 [129].

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

75

Fig. 13 PTP fluorescent A3 AR antagonists

Finally, a commercially available preladenant-based fluorescent ligand, SCH-red, has been used by Ciruela and colleagues to demonstrate the existence of native D2R-A2A AR oligomers in time-resolved fluorescence resonance energy transfer (TR-FRET) experiments, in striatal rat membranes both in normal conditions and in a Parkinson’s disease model. Studies demonstrated a decreased D2R-A2A AR oligomer formation in the 6-OHDA-lesioned striatal membranes [185]. Due to the well-known SAR of PTP on the A3 AR, different attempts to obtain potent and selective A3 AR fluorescent antagonists have been conducted by inserting the linker, and thus the fluorophore (e.g., fluorescein isothiocyanate, FITC), at the 5 position of the 8-methyl-PTP scaffold [186–188]. In a first attempt, simple alkyldiamino moieties were directly introduced at the 5 position as linkers, leading to the low affinity and mostly dual A2A AR and A3 AR ligands 78–83 (Fig. 13). Computational studies tentatively ascribed the lack of selectivity towards A3 AR to the presence of low significant hydrophobic interactions in the binding cleft [186, 187]. For this reason, authors extended the pharmacophoric portion of PTP, using as starting compound not the simple 5-amino-8-methyl-PTP but compound 84, where the benzylamido moiety confers high affinity and selectivity at the A3 AR and the carboxylic acid at the para position of the phenyl moiety represents the anchoring point for the linker [188]. A firm increase in affinity was obtained in compounds

76

F. Prencipe et al.

85–89 (Fig. 13), reaching a Ki value around 100 nM towards A3 AR independently from the linker length. Surprisingly, a rise in affinity was observed also against A2A AR, thus defining these compounds as dual fluorescent antagonists. Finally, a series of alkyne derivatives of PTP was synthetized with the aim to use the classical click chemistry approach to introduce the fluorophore (e.g., as azide derivative). The formed triazole is an aromatic ring that, when present at the 5 position, is known to give favorable interactions at the A3 AR subtype. Alkyne derivative with a length of 6 carbon atoms has been used to make the conjugate with an azido derivative of Alexa Fluor 488, leading to MRS5763 (90) that is the best A3 AR fluorescent ligand bearing a PTP scaffold, exhibiting a Ki of 31.8 nM and a 2.8-fold selectivity against A2A AR.

3.3.2

Other Conjugates

PTPs were used also to design other kind of conjugates with different purposes, such as biotin conjugate, that, thanks to its binding to streptavidin, could be used to set up a variety of experiments both for screening, receptor detection or imaging purposes (Fig. 14). Specifically, compound 91 was developed on the Scheme 442416 (5) structure, thus as tool of the A2A AR. In the same work, Kumar et al. reported dendrimers functionalized again with two different amino derivatives of Scheme 442416 (92,93). In particular, polyamidoamine (PAMAM) G3.5 dendrimer 15 (containing a butane core), that possesses peripheral carboxylic acids, has been used [182]. ARs are present on the cell surface as multi homomeric or heteromeric (with other GPCRs) entities, thus the interest in creation of multivalent confined structures able to interact with them [189]. Dendrimers 92 and 93 (Fig. 14) were highly potent and selective towards A2A AR, with practically no difference between two linkers in terms of affinity at the target receptor [182]. With the same objective to develop multivalent structures, an A3 AR antagonist has been conjugated with magnetic carbon nanotubes. Because A3 AR has been found to be overexpressed in different cancer cell lines, the magnetic property of these Fe-filled carbon nanotubes could be useful to perform cancer magnetic cell sorting and thermal therapy. The final nanostructures Fe-filled-CNTs-HEG-PTP (94) and Fe-filled-CNTs-TEG-PTP (95), differing for the used linker, have shown to compete with A3 AR radioligand in a concentration-dependent manner, but structure 94 failed to selectively target hA3 AR overexpressing cells. The problem could be ascribed to electrostatic interactions between the unreacted free amino groups on the carbon nanotubes and anionic residue on the cell membranes [190]. PTP were also implied to develop multitarget ligands, in particular a dual ligand targeting A2A AR and Histone Deacetylase (HDAC) as a possible therapeutic strategy in cancer immunotherapy. Structural features of A2A AR antagonists and HDAC inhibitors were combined thanks to a structure-based drug design leading to the discovery of IHCH-3064 (96), a PTP derivative exhibiting a Ki value towards A2A AR of 2.2 nM and an IC50 value towards HDAC of 80.2 nM. IHCH-3064 (96) showed good antiproliferative activity against tumor cell lines in vitro and, when administered intraperitoneally in mice, inhibited tumor growth [191].

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

77

Fig. 14 Other PTP conjugates

4 Conclusions ARs, and in particular A2A ARs, are among the most studied GPCRs, particularly from a structural point of view. For this reason, they represent a model for other GPCRs and several pharmacological tools and probes have been developed over the years. Among them, PTP-based antagonists were extensively investigated both at the A2A and A3 ARs. In fact, in addition to a very complete and distinct SAR profile to both receptors, PTP antagonists have been used as skeleton to develop more

78

F. Prencipe et al.

complex structures bearing specific functions. These range from simple covalent interaction or radiolabeling to conjugation to linkers in order to obtain multipurpose molecules such as fluorescent, multitarget, and/or multivalent ligands. Thus, PTP represent the perfect example on how it is possible to play with a known GPCR class of ligands in order to increase knowledge on the GPCR itself, crucial to discover new pathophysiological implications of the protein and, consequently, to develop new valuable therapies. Funding The authors have no conflicts of interest to declare that are relevant to the content of this chapter. Ethical Approval Ethical approval is not applicable for this article. Informed Consent There are no human subjects in this work and informed consent is not applicable.

References 1. Borea PA, Gessi S, Merighi S et al (2018) Pharmacology of adenosine receptors: the state of the art. Physiol Rev 98:1591–1625 2. Pearson JD (1985) Ectonucleotidases. Measurement of activities and use of inhibitors. In: Paton DM (ed) Methods in pharmacology. Methods used in adenosine research, vol 6. Plenum Press, New York, pp 83–107 3. Ontyd J, Schrader J (1984) Measurement of adenosine, inosine, and hypoxanthine in human plasma. J Chromatogr 307:404–409 4. Kostopoulos GK (1988) Adenosine: a molecule for synaptic homeostasis? Evolution of current concepts on the physiological and pathophysiological roles of adenosine in the brain. In: Avoli M, Reader TA, Dykes RW, Gloor P (eds) Neurotransmitters and cortical function: from molecules to mind. Plenum Press, New York, pp 415–435 5. Dragunow M, Faull RLM (1988) Neuroprotective effects of adenosine. Trends Pharmacol Sci 9:193–194 6. Sebastiao AM, Ribeiro JA (2000) Fine-tuning neuromodulation by adenosine. Trends Pharmacol Sci 21:341–346 7. Burnstock G (1978) A basis for distinguishing two types of purinergic receptor. In: Bolis L, Straub RW (eds) Cell membrane receptors for drugs and hormones. Raven Press, New York, pp 107–118 8. Ijzerman AP, Jacobson KA, Muller CE et al (2022) International union of basic and clinical pharmacology. CXII: a further update. Pharmacol Rev 74:340–372 9. Jacobson KA, Gao ZG (2006) Adenosine receptor as therapeutic target. Nat Rev Drug Discov 5:247–264 10. Effendi WI, Nagano T, Kobayashi K et al (2020) Focusing on adenosine receptors as a potential targeted therapy in human diseases. Cell 9:785 11. Obiefuna PCM, Batra VK, Nadeem A et al (2005) A novel A1 adenosine receptor antagonist, L-97-1 [3-[2-(4-aminophenyl)-ethyl]-8-benzyl-7-{2-ethyl-(2-hydroxy-ethyl)-amino-ethyl}-1propyl-3,7-dihydro-purine-2,6-dione], reduces allergic responses to house dust mite in an allergic rabbit model of asthma. J Pharmacol Exp Ther 315:329–336 12. Chunn JL, Young HW, Banerjee SK et al (2001) Adenosine-dependent airway inflammation and hyper responsiveness in partially adenosine deaminase-deficient mice. J Immunol 167: 4676–4685

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

79

13. Ngamsri KC, Wagner R, Vollmer I et al (2010) Adenosine receptor A1 regulates polymorphonuclear cell tracking and microvascular permeability in lipopolysaccharide-induced lung injury. J Immunol 185:4374–4384 14. Fernandez LG, Sharma AK, Lapar DJ et al (2013) Adenosine A1 receptor activation attenuates lung ischemia-reperfusion injury. J Thorac Cardiovasc Surg 145:1654–1659 15. Affner F, Woods PS, Davis IC (2014) Activation of A1-adenosine receptors promotes leukocyte recruitment to the lung and attenuates acute lung injury in mice infected with influenza A/WSN/33 (H1N1) virus. J Virol 88:10214–10227 16. Blackburn MR, Vance CO, Morschl E, Wilson CN (2009) Adenosine receptors and inflammation. In: Wilson CN, Mustafa SJ (eds) Adenosine receptors in health and disease. Springer, Berlin, pp 215–269 17. Cronstein BN, Levin RI, Philips M et al (1992) Neutrophil adherence to endothelium is enhanced via adenosine A1 receptors and inhibited via adenosine A2 receptors. J Immunol 148:2201–2206 18. Gessi S, Merighi S, Fazzi D et al (2011) Adenosine receptor targeting in health and disease. Expert Opin Investig Drugs 20:1591–1609 19. Headrick JP, Peart JN, Reichelt ME et al (2011) Adenosine and its receptors in the heart: regulation, retaliation and adaptation. Biochim Biophys Acta Biomembr 1808:1413–1428 20. Jacobson KA, Tosh DK, Jain S et al (2019) Historical and current adenosine receptor agonists in preclinical and clinical development. Front Cell Neurosci 13:124 21. Ellenbogen KA, O’Neill G, Prystowsky EN et al (2005) Trial to evaluate the management of paroxysmal supraventricular tachycardia during an electrophysiology study with tecadenoson. Circulation 111:3202–3208 22. Mason PK, DiMarco JP (2009) New pharmacological agents for arrhythmias. Circ Arrhythm Electrophysiol 2:588–597 23. Szentmiklosi A, Galajda Z, Cseppento A et al (2015) The Janus face of adenosine: antiarrhythmic and proarrhythmic actions. Curr Pharm Des 21:965–976 24. Wu LG, Saggau P (1997) Presynaptic inhibition of elicited neurotransmitter release. Trends Neurosci 20:204–212 25. Stone TW, Ceruti S, Abbracchio MP (2009) Adenosine receptors and neurological disease: neuroprotection and neurodegeneration. In: Wilson CN, Mustafa SJ (eds) Adenosine receptors in health and disease, vol 193. Springer, Berlin, pp 535–587 26. Borea PA, Gessi S, Merighi S et al (2016) Adenosine as a multi-signalling Guardian angel in human diseases: when, where and how does it exert its protective effects? Trends Pharmacol Sci 37:419–434 27. Luongo L, Petrelli R, Gatta L et al (2012) 5’-Chloro-5′-deoxy-(±)-ENBA, a potent and selective adenosine A1 receptor agonist, alleviates neuropathic pain in mice through functional glial and microglial changes without affecting motor or cardiovascular functions. Molecules 17:13712–13726 28. Fedele DE, Li T, La JQ et al (2006) Adenosine A1 receptors are crucial in keeping an epileptic focus localized. Exp Neurol 200:184–190 29. Sebastião AM, Ribeiro JA (2009) Adenosine receptors and the central nervous system. In: Wilson CN, Mustafa SJ (eds) Adenosine receptors in health and disease, vol 193. Springer, Berlin, pp 471–534 30. Peleli M, Carlstrom M (2017) Adenosine signaling in diabetes mellitus and associated cardiovascular and renal complications. Mol Aspects Med 55:62–74 31. Kiesman WF, Elzein E, Zablocki J (2009) A1 adenosine receptor antagonists, agonists, and allosteric enhancers. In: Wilson CN, Mustafa SJ (eds) Adenosine receptors in health and disease, vol 193. Springer, Berlin, pp 25–58 32. Hasko G, Pacher P (2008) A2A receptors in inflammation and injury: lessons learned from transgenic animals. J Leukoc Biol 83:447–455

80

F. Prencipe et al.

33. Wilson CN, Nadeem A, Spina D et al (2009) Adenosine receptors and asthma. In: Wilson CN, Mustafa SJ (eds) Adenosine receptors in health and disease, vol 193. Springer, Berlin, pp 329–362 34. Golzar Y, Doukky R (2014) Regadenoson use in patients with chronic obstructive pulmonary disease: the state of current knowledge. Int J COPD 9:129–137 35. Boknik P, Drzewiecki K, Eskandar J et al (2019) Evidence for arrhythmogenic effects of A2Aadenosine receptors. Front Pharmacol 10:1051 36. Dunwiddie TV, Masino SA (2001) The role and regulation of adenosine in the central nervous system. Annu Rev Neurosci 24:31–55 37. Boison D, Singer P, Shen HY et al (2012) Adenosine hypothesis of schizophreniaopportunities for pharmacotherapy. Neuropharmacology 62:1527–1543 38. Loram LC, Harrison JA, Sloane EM et al (2009) Enduring reversal of neuropathic pain by a single intrathecal injection of adenosine A2A receptor agonists: a novel therapy for neuropathic pain. J Neurosci 29:14015–14025 39. Knezevic NN, Cicmil N, Knezevic I et al (2015) Discontinued neuropathic pain therapy between 2009-2015. Expert Opin Investig Drugs 24:1631–1646 40. Knapp CM, Foye MM, Cottam N et al (2001) Adenosine agonists CGS 21680 and NECA inhibit the initiation of cocaine self-administration. Pharmacol Biochem Behav 68:797–803 41. Mohamed RA, Agha AM, Abdel-Rahman AA et al (2016) Role of adenosine A2A receptor in cerebral ischemia reperfusion injury: signaling to phosphorylated extracellular signalregulated protein kinase (pERK1/2). Neuroscience 314:145–159 42. Yamada K, Kobayashi M, Kanda T (2014) Involvement of adenosine A2A receptors in depression and anxiety. Int Rev Neurobiol 119:373–393 43. Faivre E, Coelho JE, Zornbach K et al (2018) Beneficial effect of a selective adenosine A2A receptor antagonist in the APPswe/PS1dE9 mouse model of Alzheimer’s disease. Front Mol Neurosci 11:235 44. Popoli P, Blum D, Martire A et al (2007) Functions, dysfunctions and possible therapeutic relevance of adenosine A2A receptors in Huntington’s disease. Prog Neurobiol 81:331–348 45. De Lera RM, Lim YH, Zheng J (2014) Adenosine A2A receptor as a drug discovery target. J Med Chem 57:3623–3650 46. Nazario LR, da Silva RS, Bonan CD (2017) Targeting adenosine signaling in Parkinson’s disease: from pharmacological to non-pharmacological approaches. Front Neurosci 11:658 47. Ribeiro JA, Sebastião AM, De Mendonça A (2002) Adenosine receptors in the nervous system: pathophysiological implications. Prog Neurobiol 68:377–392 48. Li X, Kang H, Liu X et al (2012) Effect of adenosine A2A receptor antagonist ZM241385 on amygdala-kindled seizures and progression of amygdala kindling. J Huazhong Univ Sci Technolog Med Sci 32:257–264 49. Sek K, Mølck C, Stewart GD et al (2018) Targeting adenosine receptor signaling in cancer immunotherapy. Int J Mol Sci 19:3837 50. Congreve M, Brown GA, Borodovsky A et al (2018) Targeting adenosine A2A receptor antagonism for treatment of cancer. Expert Opin Drug Discovery 13:997–1003 51. Antonioli L, Csoka B, Fornai M et al (2014) Adenosine and inflammation: what’s new on the horizon? Drug Discov Today 19:1051–1068 52. Linden J (2006) New insights into the regulation of inflammation by adenosine. J Clin Investig 116:1835–1837 53. Gao ZG, Balasubramanian R, Kiselev E et al (2014) Probing biased/partial agonism at the G protein-coupled A2B adenosine receptor. Biochem Pharmacol 90:297–306 54. Van Den Berge M, Hylkema MN, Versluis M et al (2007) Role of adenosine receptors in the treatment of asthma and chronic obstructive pulmonary disease: recent developments. Drugs R D 8:13–23 55. Mustafa SJ, Nadeem A, Fan M et al (2007) Effect of a specific and selective A2B adenosine receptor antagonist on adenosine agonist AMP and allergen-induced airway responsiveness and cellular influx in a mouse model of asthma. J Pharmacol Exp Ther 320:1246–1251

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

81

56. Eckle T, Grenz A, Laucher S et al (2008) A2B adenosine receptor signaling attenuates acute lung injury by enhancing alveolar fluid clearance in mice. J Clin Investig 118:3301–3315 57. Hoegl S, Brodsky KS, Blackburn MR et al (2015) Alveolar epithelial A2B adenosine receptors in pulmonary protection during acute lung injury. J Immunol 195:1815–1824 58. Haskó G, Csóka B, Németh ZH et al (2009) A2B adenosine receptors in immunity and inflammation. Trends Immunol 30:263–270 59. Maas JE, Wan TC, Figler RA et al (2010) Evidence that the acute phase of ischemic preconditioning does not require signaling by the A2B adenosine receptor. J Mol Cell Cardiol 49:886–893 60. Dubey RK, Gillespie DG, Osaka K et al (1996) Adenosine inhibits growth of rat aortic smooth muscle cells. Possible role of A2B receptor. Hypertension 27:786–793 61. Eisenstein A, Patterson S, Ravid K (2015) The many faces of the A2B adenosine receptor in cardiovascular and metabolic diseases. J Cell Physiol 230:2891–2897 62. Németh ZH, Bleich D, Csóka B et al (2007) Adenosine receptor activation ameliorates type 1 diabetes. FASEB J 21:2379–2388 63. Sun Y, Huang P (2016) Adenosine A2B receptor: from cell biology to human diseases. Front Chem 4:37 64. Cekic C, Sag D, Li Y et al (2012) Adenosine A2B receptor blockade slows growth of bladder and breast tumors. J Immunol 188:198–205 65. Walker BA, Jacobson MA, Knight DA et al (1997) Adenosine A3 receptor expression and function in eosinophils. Am J Respir Cell Mol Biol 16:531–537 66. Gao Z, Li BS, Day YJ et al (2001) A3 adenosine receptor activation triggers phosphorylation of protein kinase B and protects rat basophilic leukemia 2H3 mast cells from apoptosis. Mol Pharmacol 59:76–82 67. Martin L, Pingle SC, Hallam DM et al (2006) Activation of the adenosine A3 receptor in RAW 264.7 cells inhibits lipopolysaccharide-stimulated tumor necrosis factor-alpha release by reducing calcium-dependent activation of nuclear factor-kappaB and extracellular signalregulated kinase 1/2. J Pharmacol Exp Ther 316:71–78 68. Lee JY, Jhun BS, Oh YT et al (2006) Activation of adenosine A3 receptor suppresses lipopolysaccharide-induced TNF-alpha production through inhibition of PI 3-kinase/Akt and NF-kappaB activation in murine BV2 microglial cells. Neurosci Lett 396:1–6 69. Cohen S, Barer F, Bar-Yehuda S et al (2014) A3 adenosine receptor allosteric modulator induces an anti-inflammatory effect: in vivo studies and molecular mechanism of action. Mediators Inflamm 3:708746 70. David M, Akerman L, Ziv M et al (2012) Treatment of plaque-type psoriasis with oral CF101: data from an exploratory randomized phase 2 clinical trial. J Eur Acad Dermatol Venereol 26: 361–367 71. Yang H, Avila MY, Peterson-Yantorno K et al (2005) The cross-species A3 adenosinereceptor antagonist MRS 1292 inhibits adenosine-triggered human nonpigmented ciliary epithelial cell fluid release and reduces mouse intraocular pressure. Curr Eye Res 30:747–754 72. Okamura T, Kurogi Y, Hashimoto K et al (2004) Structure-activity relationships of adenosine A3 receptor ligands: new potential therapy for the treatment of glaucoma. Bioorg Med Chem Lett 14:3775–3779 73. Zhong H, Shlykov SG, Molina JG et al (2003) Activation of murine lung mast cells by the adenosine A3 receptor. J Immunol 171:338–345 74. Borea PA, Varani K, Vincenzi F et al (2015) The A3 adenosine receptor: history and perspectives. Pharmacol Rev 67:74–102 75. Headrick JP, Lasley RD (2009) Adenosine receptors and reperfusion injury of the heart. In: Wilson CN, Mustafa SJ (eds) Adenosine receptors in health and disease, vol 193. Springer, Berlin, pp 189–214 76. Auchampach JA, Ge ZD, Wan TC et al (2003) A3 adenosine receptor agonist IB-MECA reduces myocardial ischemia-reperfusion injury in dogs. Am J Physiol Heart Circ Physiol 285: H607–H613

82

F. Prencipe et al.

77. Ge ZD, Peart JN, Kreckler LM et al (2006) Cl-IB-MECA [2-chloro-N6-(3-iodobenzyl)adenosine-5’-N-methylcarboxamide] reduces ischemia/reperfusion injury in mice by activating the A3 adenosine receptor. J Pharmacol Exp Ther 319:1200–1210 78. Black RGJ, Guo Y, Ge ZD et al (2002) Gene dosage-dependent effects of cardiac-specific overexpression of the A3 adenosine receptor. Circ Res 91:165–172 79. Park JG, Jeong SJ, Yu J et al (2018) LJ-1888, a selective antagonist for the A3 adenosine receptor, ameliorates the development of atherosclerosis and hypercholesterolemia in apolipoprotein E knock-out mice. BMB Rep 51:520–525 80. Rothermel BA, Hill JA (2008) Adenosine A3 receptor and cardioprotection: enticing, enigmatic, elusive. Circulation 118:1691–1693 81. Pugliese AM, Coppi E, Volpini R et al (2007) Role of adenosine A3 receptors on CA1 hippocampal neurotransmission during oxygen-glucose deprivation episodes of different duration. Biochem Pharmacol 74:768–779 82. Wittendorp MC, Boddeke HWGM, Biber K (2004) Adenosine A3 receptor-induced CCL2 synthesis in cultured mouse astrocytes. Glia 46:410–418 83. Etherington LAV, Frenguelli BG (2004) Endogenous adenosine modulates epileptiform activity in rat hippocampus in a receptor subtype-dependent manner. Eur J Neurosci 19: 2539–2550 84. Durante M, Squillace S, Lauro F et al (2021) Adenosine A3 agonists reverse neuropathic pain via T cell-mediated production of IL10. J Clin Invest 131:e139299 85. Roseti C, Palma E, Martinello K et al (2009) Blockage of A2A and A3 adenosine receptors decreases the desensitization of human GABAA receptors microtransplanted to Xenopus oocytes. Proc Natl Acad Sci U S A 106:15927–15931 86. Mabley J, Soriano F, Pacher P et al (2003) The adenosine A3 receptor agonist, N6-(3-iodobenzyl)-adenosine-5’-N-methyluronamide, is protective in two murine models of colitis. Eur J Pharmacol 466:323–329 87. Lee HT, Ota-Setlik A, Xu H et al (2003) A3 adenosine receptor knockout mice are protected against ischemia- and myoglobinuria-induced renal failure. Am J Physiol Renal Physiol 284: F267–F273 88. Madi L, Ochaion A, Rath-Wolfson L et al (2004) The A3 adenosine receptor is highly expressed in tumor versus normal cells: potential target for tumor growth inhibition. Clin Cancer Res 10:4472–4479 89. Gessi S, Merighi S, Varani K et al (2008) The A3 adenosine receptor: an enigmatic player in cell biology. Pharmacol Ther 117:123–140 90. Fishman P, Bar-Yehuda S, Vagman L (1998) Adenosine and other low molecular weight factors released by muscle cells inhibit tumor cell growth. Cancer Res 58:3181–3187 91. Fishman P, Bar-Yehuda S, Ohana G et al (2000) Adenosine acts as an inhibitor of lymphoma cell growth: a major role for the A3 adenosine receptor. Eur J Cancer 36:1452–1458 92. Zhang X, Zhang M, Laties AM et al (2006) Balance of purines may determine life or death of retinal ganglion cells as A3 adenosine receptors prevent loss following P2X7 receptor stimulation. J Neurochem 98:566–575 93. Gessi S, Merighi S, Varani K et al (2007) Adenosine receptors in colon carcinoma tissues and colon tumoral cell lines: focus on the A3 adenosine subtype. J Cell Physiol 211:826–836 94. Ohana G, Bar-Yehuda S, Arich A et al (2003) Inhibition of primary colon carcinoma growth and liver metastasis by the A3 adenosine receptor agonist CF101. Br J Cancer 89:1552–1558 95. Madi L, Bar-Yehuda S, Barer F et al (2003) A3 adenosine receptor activation in melanoma cells: association between receptor fate and tumor growth inhibition. J Biol Chem 278:42121– 42130 96. Fishman P, Bar-Yehuda S, Ardon E et al (2003) Targeting the A3 adenosine receptor for cancer therapy: inhibition of prostate carcinoma cell growth by A3AR agonist. Anticancer Res 23:2077–2083 97. Francis JE, Cash WD, Psychoyos S et al (1988) Structure-activity profile of a series of novel triazoloquinazoline adenosine antagonists. J Med Chem 31:1014–1020

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

83

98. Sarges R, Howard HR, Browne RG et al (1990) 4-Amino[1,2,4]triazolo[4,3-a]quinoxalines. A novel class of potent adenosine receptor antagonists and potential rapid-onset antidepressants. J Med Chem 33:2240–2254 99. Jacobson KA, van Galen PJM, Williams M (1992) Adenosine receptors: pharmacology, structure-activity relationships, and therapeutic potential. J Med Chem 35:407–422 100. Gatta F, Del Giudice MR, Borioni A et al (1993) Synthesis of imidazo[1,2-c]pyrazolo[4,3-e] 1,2,4-triazolo[1,5-c]pyrimidines and 1,2,4-triazolo[5,1-i]purines: new potent adenosine A2 receptor antagonists. Eur J Med Chem 28:569–576 101. Baraldi PG, Manfredini S, Simoni D et al (1994) Synthesis of new pyrazolo[4,3-e]1,2,4triazolo[1,5-c] pyrimidine and 1,2,3-triazolo[4,5-e]1,2,4-triazolo[1,5-c] pyrimidine displaying potent and selective activity as A2A adenosine receptor antagonists. Bioorg Med Chem Lett 4: 2539–2544 102. Baraldi PG, Cacciari B, Spalluto G et al (1996) Pyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidine derivatives: potent and selective A2A adenosine antagonists. J Med Chem 39:1164–1171 103. Ongini E (1997) SCH 58261: a selective A2A adenosine receptor antagonists. Drug Dev Res 42:63–70 104. Baraldi PG, Cacciari B, Spalluto G et al (1998) Design, synthesis and biological evaluation of a second generation of pyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidines as potent and selective A2A adenosine receptor antagonists. J Med Chem 41:2126–2133 105. Baraldi PG, Cacciari B, Romagnoli R et al (1999) Pyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidines derivatives as highly potent and selective human A3 adenosine receptor antagonists. J Med Chem 42:4473–4478 106. Baraldi PG, Cacciari B, Moro S et al (2001) Fluorosulfonyl- and bis-(β-chloroethyl)aminophenylamino functionalized pyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidines derivatives: irreversible antagonists at the human A3 adenosine receptor and molecular modelling studies. J Med Chem 44:2735–2742 107. Baraldi PG, Cacciari B, Romagnoli R et al (2002) 7-Substituted 5-Amino-2-(2-furyl)pyrazolo [4,3-e]1,2,4-triazolo[1,5-c]pyrimidines as A2A adenosine receptor antagonists: a study on the importance of modifications at the side chain on the activity and solubility. J Med Chem 45: 115–126 108. Baraldi PG, Fruttarolo F, Tabrizi MA et al (2003) Design synthesis and biological evaluation of c9- and C2-substituted pyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidines as new A2A and A3 adenosine receptor antagonists. J Med Chem 46:1229–1241 109. Neustadt BR, Hao J, Lindo N et al (2007) Potent, selective and orally active adenosine A2A receptor antagonists: arylpiperazine derivatives of pyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidines. Bioorg Med Chem Lett 17:1376–1380 110. Cheong SL, Dolzhenko A, Kachler S et al (2010) The significance of 2-furyl ring substitution with a 2-(para-substituted) aryl group in a new series of pyrazolo-triazolo-pyrimidines as potent and highly selective hA3 adenosine receptors antagonists: new insights into structureaffinity relationship and receptor-antagonist recognition. J Med Chem 53:3361–3375 111. Harris JM, Neustadt BR, Zhang H et al (2011) Potent and selective adenosine A2A receptor antagonists: [1,2,4]-triazolo[4,3-c]pyrimidin-3-ones. Bioorg Med Chem Lett 21:2497–2501 112. Baraldi PG, Saponaro G, Romagnoli R et al (2012) Water-soluble pyrazolo[4,3-e][1,2,4] triazolo[1,5-c]pyrimidines as human A3 adenosine receptor antagonists. J Med Chem 55: 5380–5390 113. Federico S, Redenti S, Sturlese M et al (2015) The influence of the 1-(3-trifluoromethylbenzyl)-1H-pyrazole-4-yl moiety on the adenosine receptors affinity profile of pyrazolo[4,3-e] [1,2,4]triazolo[1,5-c]pyrimidine derivatives. PloS One 10:e0143504 114. Redenti S, Ciancetta A, Pastorin G et al (2016) Pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidines and structurally simplified analogs. Chemistry and SAR profile as adenosine receptor antagonists. Curr Top Med Chem 16:3224–3257 115. Hodgson RA, Bertorelli R, Varty GB et al (2009) Characterization of the potent and highly selective A2A receptor antagonist preladenant and SCH 412348 [7-[2-[4-2,4-difluorophenyl]-

84

F. Prencipe et al.

1-piperazinyl]ethyl-2-(2-furanyl)-7H-pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5amine] in rodent models of movement disorders and depression. J Pharmacol Exp Ther 330: 294–303 116. Hauser R, Stocchi F, Rascol O et al (2014) Phase-3 clinical trials of adjunctive therapy with preladenant, an adenosine 2a antagonist, in patient with Parkinson’s disease. Neurology 82: P7.087 117. Stocchi F, Rascol O, Hauser R et al (2014) Phase-3 clinical trials of adjunctive therapy with preladenant, an adenosine 2a antagonist, in patient with Parkinson’s disease. Neurology 82 (S7):004 118. Zocchi C, Ongini E, Conti A et al (1996) The non-xanthine heterocyclic compound SCH58261 is a new potent and selective A2A adenosine receptor antagonist. J Pharmacol Exp Ther 276: 398–404 119. Fredholm BB, Lindstrom K, Dionisotti S et al (1998) [3H]-SCH58261, a selective adenosine A2A receptor antagonist, is a useful ligand in autoradiographic studies. J Neurochem 70:1210– 1216 120. Svenningsson P, Nomikos GG, Ongini E et al (1997) Antagonism of adenosine A2A receptors underlies the behavioural activating effect of caffeine and is associated with reduced expression of mRNA for NGFI-A and NGFI-B in caudate-putamen and nucleus accumbens. Neuroscience 79:753–764 121. Bertorelli R, Ferri N, Adami M et al (1996) Effects of selective agonists and antagonists for A1 and A2A adenosine receptors on sleep-waking patterns in rats. Drug Dev Res 37:65–72 122. Monopoli A, Casati C, Lozza G et al (1998) Cardiovascular pharmacology of the A2A adenosine receptor antagonist, SCH 58261, in the rat. J Pharmacol Exp Ther 285:9–15 123. Wardas J (2003) Synergistic effect of SCH 58261, an adenosine A2A receptor antagonist, and L-DOPA on the reserpine-induced muscle rigidity in rats. Pol J Pharmacol 55:155–164 124. Wardas J, Konieczny J, Lorenc-Koci E (2001) SCH 58261, an A2A adenosine receptor antagonist, counteracts parkinsonian-like muscle rigidity in rats. Synapse 41:160–171 125. Yong L, Xiaohong L, Shouyue H et al (2015) Effect of SCH442416 on glutamate uptake in retinal Muller cells at increased hydrostatic pressure. Mol Med Rep 12:3993–3997 126. Orrú M, Quiroz C, Guitart X et al (2011) Pharmacological evidence for different populations of postsynaptic adenosine A2A receptors in the rat striatum. Neuropharmacology 61:967–974 127. Baraldi PG, Borea PA (2000) New potent and selective human adenosine A3 receptor antagonists. Trends Pharmacol Sci 21:456–459 128. Varani K, Merighi S, Gessi S et al (2000) [3H]MRE 3008F20: a novel antagonist radioligand for the pharmacological and biochemical characterization of human A3 adenosine receptors. Mol Pharmacol 57:968–975 129. Claff T, Klapschinski TA, Tiruttani Subhramanyam UK et al (2022) Single stabilizing point mutation enables high-resolution co-crystal structures of the adenosine A2A receptor with preladenant conjugates. Angew Chem Int Ed 61:e202115545 130. Sajjadi FG, Firestein GS (1993) cDNA cloning and sequence analysis of the human A3 adenosine receptor. Biochim Biophys Acta 1179:105–107 131. Salvatore CA, Jacobson MA, Taylor HE (1993) Molecular cloning and characterization of the human A3 adenosine receptor. Proc Natl Acad Sci U S A 90:10365–10369 132. Linden J, Taylor HE, Robeva AS (1993) Molecular cloning and functional expression of a sheep A3 adenosine receptor with widespread tissue distribution. Mol Pharmacol 44:524–532 133. Zhao ZH, Ravid S, Ravid K (1995) Chromosomal mapping of the mouse A3 adenosine receptor gene, adora3. Genomics 30:118–119 134. Hill RJ, Oleynek JJ, Hoth CF (1997) Cloning, expression and pharmacological characterization of rabbit A1 and A3 receptors. J Pharmacol Exp Ther 280:122–128 135. Muller CE (2003) Medicinal chemistry of adenosine A3 receptor ligands. Curr Top Med Chem 3:445–462

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

85

136. Kim YC, Ji XD, Jacobson KA (1996) Derivatives of the triazoloquinazoline adenosine antagonist (GCS15943) are selective for the human A3 receptor subtype. J Med Chem 39: 4142–4148 137. Baraldi PG, Cacciari B, Spalluto G et al (1996) Novel N6-(substituted-phenylcarbamoyl) adenosine-5′-uronamides as potent agonists for A3 adenosine receptor. J Med Chem 39: 802–806 138. Baraldi PG, Cacciari B, Romagnoli R et al (2000) Pyrazolo[3,4-e]1,2,4-triazolo[1,5-c]pyrimidine derivatives as highly potent and selective human A3 adenosine receptor antagonists: influence of the chain at the N8 pyrazole nitrogen. J Med Chem 43:4768–4780 139. Baraldi PG, Cacciari B, Moro S et al (2002) Synthesis, biological activity and molecular modeling investigation of new pyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidine derivatives as human A3 adenosine receptor antagonists. J Med Chem 45:770–780 140. Bolcato C, Cusan C, Pastorin G et al (2008) Pyrazolo-triazolo-pyrimidines as adenosine receptor antagonists: effect of the N-5 bond type on the affinity and selectivity at the four adenosine receptor subtypes. Purinergic Signal 4:39–46 141. Maconi A, Pastorin G, Da Ros T et al (2002) Synthesis, biological properties and molecular modeling investigation of the first potent, selective and water soluble human A3 adenosine receptor antagonist. J Med Chem 45:3579–3582 142. Pastorin G, Da Ros T, Bolcato C et al (2006) Synthesis and biological studies of a new series of 5-heteroarylcarbamoylaminopyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidines as human A3 adenosine receptor antagonists. Influence of the heteroaryl substituent on binding affinity and molecular modeling investigation. J Med Chem 49:1720–1729 143. Federico S, Ciancetta A, Sabbadin D et al (2012) Exploring the directionally of 5-substitutions in a new series of 5-alkylaminopyrazolo[4,3-e]1,2,4-triazolo[1,5-c]pyrimidine as a strategy to design novel human A3 adenosine receptor antagonists. J Med Chem 55:9654–9668 144. Federico S, Margiotta E, Moro S et al (2021) Potent and selective A3 adenosine receptor antagonists bearing aminoesters as heterobifunctional moieties. RCS Med Chem 12:254–262 145. Cheong SL, Federico S, Venkatesan G et al (2013) The A3 adenosine receptor as multifaceted therapeutic target: pharmacology, medicinal chemistry and in silico approaches. Med Res Rev 33:235–335 146. Baraldi PG, Cacciari B, Romagnoli R et al (2001) Pyrazolo[3,4-e]1,2,4-triazolo[1,5-c]pyrimidine derivatives as adenosine receptor ligands: a starting point for searching A2B adenosine receptor antagonists. Drug Res Rev 53:225–235 147. Pastorin G, Da Ros T, Spalluto G et al (2003) Pyrazolo[3,4-e]1,2,4-triazolo[1,5-c]pyrimidine derivatives as adenosine receptor antagonists. Influence of the N5 substituent on the affinity at the human A3 and A2B adenosine receptor subtypes: a molecular modeling investigation. J Med Chem 46:4287–4296 148. Michielan L, Federico S, Terfloth L et al (2009) Exploring potency and selectivity receptor antagonist profiles using a multilabel classification approach: the human adenosine receptors as a key study. J Chem Inf Model 49:2820–2286 149. Beauglehole AR, Baker SP, Scammells PJ (2002) New irreversible adenosine A1 antagonists based on FSCPX. Bioorg Med Chem Lett 12:3179–3182 150. Glukhova A, Thal DM, Nguyen AT et al (2017) Structure of the adenosine A1 receptor reveals the basis for subtype selectivity. Cell 168:867–877.e13 151. Weichert D, Gmeiner P (2015) Covalent molecular probes for class A G protein-coupled receptors: advances and applications. ACS Chem Biol 10:1376–1386 152. Shryock JC, Snowdy S, Baraldi PG et al (1998) A2A-adenosine receptor reserve for coronary vasodilation. Circulation 98:711–718 153. Grunbeck A, Sakmar TP (2013) Probing G protein-coupled receptor—ligand interactions with targeted photoactivatable cross-linkers. Biochemistry 52:8625–8632 154. Muranaka H, Momose T, Handa C, Ozawa T (2017) Photoaffinity labeling of the human A2A adenosine receptor and cross-link position analysis by mass spectrometry. ACS Med Chem Lett 8:660–665

86

F. Prencipe et al.

155. Matthews PM, Rabiner EA, Passchier J, Gunn RN (2012) Positron emission tomography molecular imaging for drug development. Br J Clin Pharmacol 73:175–186 156. Honer M, Gobbi L, Martarello L, Comley RA (2014) Radioligand development for molecular imaging of the central nervous system with positron emission tomography. Drug Discov Today 19:1936–1944 157. Baraldi PG, Cacciari B, Dionisotti S et al (1996) Synthesis of the tritium labeled SCH 58261, a new non-xanthine A2A adenosine receptor antagonist. J Label Compd Radiopharm 38:725– 732 158. Dionisotti S, Ongini E, Zocchi C et al (1997) Characterization of human A2A adenosine receptors with the antagonist radioligand [3H]-SCH 58261. Br J Pharmacol 121:353–360 159. El Yacoubi M, Ledent C, Parmentier M et al (2001) In vivo labelling of the adenosine A2A receptor in mouse brain using the selective antagonist [3H]SCH 58261. Eur J Neurosci 14: 1567–1570 160. Svenningsson P, Hall H, Gö G et al (1997) Distribution of adenosine receptors in the postmortem human brain: an extended autoradiographic study. Synapse 27:322–335 161. Zocchi C, Ongini E, Ferrara S et al (1996) Binding of the radioligand [3H]-SCH 58261, a new non-xanthine A2A adenosine receptor antagonist, to rat striatal membranes. Br J Pharmacol 117:1381–1386 162. Bryant R, McGuinness D, Turek-Etienne T et al (2004) WGA-coated yttrium oxide beads enable an imaging-based adenosine 2A receptor binding scintillation proximity assay suitable for high throughput screening. Assay Drug Dev Technol 2:290–299 163. Todde S, Moresco RM, Simonelli P et al (2000) Design, radiosynthesis, and biodistribution of a new potent and selective ligand for in vivo imaging of the adenosine A2A receptor system using positron emission tomography. J Med Chem 43:4359–4362 164. Moresco RM, Todde S, Belloli S et al (2005) In vivo imaging of adenosine A2A receptors in rat and primate brain using [11C]SCH442416. Eur J Nucl Med Mol Imaging 32:405–413 165. Varani K, Vincenzi F, Tosi A et al (2010) A2A adenosine receptor overexpression and functionality, as well as TNF-α levels, correlate with motor symptoms in Parkinson’s disease. FASEB J 24:587–598 166. Khanapur S, Paul S, Shah A et al (2014) Development of [18F]-labeled pyrazolo[4,3-e]-1,2,4triazolo[1, 5-c]pyrimidine (SCH442416) analogs for the imaging of cerebral adenosine A2A receptors with positron emission tomography. J Med Chem 57:6765–6780 167. Bhattacharjee AK, Lang L, Jacobson O et al (2011) Striatal adenosine A2A receptor-mediated positron emission tomographic imaging in 6-hydroxydopamine-lesioned rats using [18F]MRS5425. Nucl Med Biol 38:897–906 168. Schröder S, Lai TH, Toussaint M et al (2020) PET imaging of the adenosine A2A receptor in the rotenone-based mouse model of Parkinson’s disease with [18F]FESCH synthesized by a simplified two-step one-pot radiolabeling strategy. Molecules 25:1633 169. Lai TH, Toussaint M, Teodoro R et al (2021) Improved in vivo PET imaging of the adenosine A2A receptor in the brain using [18F]FLUDA, a deuterated radiotracer with high metabolic stability. Eur J Nucl Med Mol Imaging 48:2727–2736 170. Rosenberg AJ, Cheung Y, Liu F et al (2022) Automated synthesis of (R)-[18F]MH.MZ on the iPhase Flexlab reaction platform. J Label Compd Radiopharm 65:223–229 171. Hesk D, Borges S, Dumpit R et al (2017) Synthesis of 3H, 2H4, and 14C-MK 3814 (preladenant). J Label Compd Radiopharm 60:194–199 172. Tavares AADS, Batis J, Barret O et al (2013) In vivo evaluation of [123I]MNI-420: a novel single photon emission computed tomography radiotracer for imaging of adenosine 2A receptors in brain. Nucl Med Biol 40:403–409 173. Tavares AAS, Batis JC, Papin C et al (2013) Kinetic modeling, test–retest, and dosimetry of 123 I-MNI-420 in humans. J Nucl Med 54:1760–1767 174. Barret O, Hannestad J, Alagille D et al (2014) Adenosine 2A receptor occupancy by tozadenant and preladenant in rhesus monkeys. J Nucl Med 55:1712–1718

Adenosine Receptor Ligands, Probes, and Functional Conjugates: A 20-Year. . .

87

175. Barret O, Hannestad J, Vala C et al (2015) Characterization in humans of 18F-MNI-444, a PET radiotracer for brain adenosine 2A receptors. J Nucl Med 56:586–591 176. Yang X, Heitman LH, Jzerman AP, van der Es D (2021) Molecular probes for the human adenosine receptors. Purinergic Signal 17:85–108 177. Baraldi PG, Cacciari B, Romagnoli R et al (2000) Synthesis and preliminary biological evaluation of [3H]-MRE 3008-F20: the first high affinity radioligand antagonist for the human A3 adenosine receptors. Bioorg Med Chem Lett 10:209–211 178. Gessi S, Varani K, Merighi S et al (2004) Expression of A3 adenosine receptors in human lymphocytes: up-regulation in T cell activation. Mol Pharmacol 65:711–719 179. Gessi S, Cattabriga E, Avitabile A et al (2004) Elevated expression of A3 adenosine receptors in human colorectal cancer is reflected in peripheral blood cells. Clin Cancer Res 10:5895– 5901 180. Comeo E, Kindon ND, Soave M et al (2020) Subtype-selective fluorescent ligands as pharmacological research tools for the human adenosine A2A receptor. J Med Chem 63: 2656–2672 181. Kecskés M, Kumar TS, Yoo L et al (2010) Novel Alexa Fluor-488 labeled antagonist of the A2A adenosine receptor: application to a fluorescence polarization-based receptor binding assay. Biochem Pharmacol 80:506–511. https://doi.org/10.1016/j.bcp.2010.04.027 182. Kumar TS, Mishra S, Deflorian F et al (2011) Molecular probes for the A2A adenosine receptor based on a pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine scaffold. Bioorg Med Chem Lett 21:2740–2745. https://doi.org/10.1016/j.bmcl.2010.11.082 183. Duroux R, Ciancetta A, Mannes P et al (2017) Bitopic fluorescent antagonists of the A2A adenosine receptor based on pyrazolo[4,3-e] [1,2,4]triazolo[1,5-c] pyrimidin-5-amine functionalized congeners. Med Chem Commun 8:1659–1667 184. Gao ZG, Toti KS, Campbell R et al (2020) Allosteric antagonism of the A2A adenosine receptor by a series of bitopic ligands. Cell 9:1200 185. Ciruela F, Fernández-Dueñas V, Jacobson KA (2015) Lighting up G protein-coupled purinergic receptors with engineered fluorescent ligands. Neuropharmacology 98:58–67 186. Federico S, Margiotta E, Paoletta S et al (2019) Pyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidines to develop functionalized ligands to target adenosine receptors: fluorescent ligands as an example. Med Chem Commun 10:1094–1108 187. Kozma E, Kumar TS, Federico S et al (2012) Novel fluorescent antagonist as a molecular probe in A3 adenosine receptor binding assays using flow cytometry. Biochem Pharmacol 83: 1552–1561 188. Federico S, Margiotta E, Moro S et al (2020) Conjugable A3 adenosine receptor antagonists for the development of functionalized ligands and their use in fluorescent probes. Eur J Med Chem 186:111886 189. Jacobson KA (2013) Structure-based approaches to ligands for G-protein-coupled adenosine and P2Y receptors, from small molecules to nanoconjugates. J Med Chem 56:3749–3767 190. Pineux F, Federico S, Klotz K-N et al (2020) Targeting G protein-coupled receptors with magnetic carbon nanotubes: the case of the A3 adenosine receptor. ChemMedChem 15:1909– 1920 191. Yan W, Ling L, Wu Y et al (2021) Structure-based design of dual-acting compounds targeting adenosine A2A receptor and histone deacetylase as novel tumor immunotherapeutic agents. J Med Chem 64:16573–16597

Top Med Chem (2023) 41: 89–100 https://doi.org/10.1007/7355_2022_155 © The Author(s), under exclusive license to Springer Nature Switzerland AG 2022 Published online: 25 November 2022

Adenosine Receptor Ligands as Potential Therapeutic Agents for Impaired Wound Healing and Fibrosis Flavia Varano, Daniela Catarzi, Erica Vigiani, Sara Calenda, and Vittoria Colotta

Contents 1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 The Wound Healing Process . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3 Adenosine in Wound Healing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 Adenosine and Fibrosis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5 Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

90 91 92 95 96 96

Abstract Wound healing is a complex process that involves a series of events including inflammation, tissue formation, angiogenesis, and tissue remodeling. Such events must be well coordinated with each other, and highly regulated. Otherwise, the wounds do not heal, incurring into chronic wounds, or the healing process, excessively activated, leads to fibrosis. Adenosine, a widely distributed nucleoside, is involved in every phase of the wound healing process through the activation of its receptors (A1, A2A, A2B, and A3 adenosine receptors ARs). Thus, targeting ARs may represent a new therapeutic approach to treating chronic wounds or fibrosis, the major pathologies related to impaired wound healing. Keywords Adenosine, Adenosine receptors, Chronic ulcers, Fibrosis, Wound healing

F. Varano (✉), D. Catarzi, E. Vigiani, S. Calenda, and V. Colotta Dipartimento di Neuroscienze, Psicologia, Area del Farmaco e Salute del Bambino, Sezione di Farmaceutica e Nutraceutica, Universita’degli Studi di Firenze, Florence, Italy e-mail: flavia.varano@unifi.it

90

F. Varano et al.

1 Introduction Wound healing is one of the most complicated processes that occurs in the human body and is made up of four partially overlapping phases: hemostasis, inflammation, proliferation, and remodeling [1]. For some people, the process does not work, and wounds do not heal in a fair amount of time or at all. Patients with wound healing disorders are often old and suffer from multiple co-morbidities such as diabetes and cardiovascular disease. Compromised wound healing is associated with pain, disability, loss of productiveness, and depression. Patients suffering from abnormalities of wound healing are reported to be about 8.2 million only in the USA, and the estimated annual cost to treat wound complications is around 28 billion US dollars [2, 3]. Moreover, chronic ulcers have been related to increased mortality; in fact, 5-year mortality data indicate that diabetic ischemic ulcers are more lethal than some types of cancer [4]. Despite a clear unmet clinical need, there are currently few FDA-approved therapies for the treatment of chronic ulcers such as venous leg ulcers and diabetic foot ulcers. These include becaplermin, a recombinant human platelet-derived growth factor (rhPDFGBB), and tissue-engineered living human skin substitutes, Apligraf and Dermagraft [5–7]. Although tissue repair is initially beneficial, the wound healing process may lead to excess matrix production, scarring, and fibrosis with consequent organ dysfunction [8]. Fibrosis is responsible for a wide range of clinically important diseases including idiopathic pulmonary fibrosis, liver cirrhosis, cardiovascular fibrosis, hypertrophic scarring, keloid formation, radiation fibrosis, and scleroderma [9– 14]. Scleroderma, also known as systemic sclerosis, is a rare disease that causes hardening of the skin, and it can also affect blood vessels, internal organs, and the digestive tract. There is no cure for scleroderma, available treatments can only ease symptoms, slow disease progression improving patients’ quality of life. Ionizing radiation therapy of common tumors, such as head, neck, and breast cancers, must often be stopped due to the appearance of radiation dermatitis in the short term and dermal fibrosis in the long term. Finally, disfiguring scars are a major cause of physical and psychological disability after trauma, resulting in more than 170,000 scar revision surgeries each year, with a significant healthcare cost. A variety of different factors regulate the wound healing process, ranging from growth factors to small molecules released at the wounded site. One of such molecules is adenosine, an endogenous purine nucleoside that is generated extracellularly from the serial dephosphorylation of ATP to AMP, by nucleoside triphosphate dephosphorylase (CD39), followed by dephosphorylation to adenosine by 5′-ectonucleotidase (CD73) [15]. Extracellular adenosine is then degraded to inosine by the adenosine deaminase enzyme (ADA). Adenosine mediates its pathophysiological actions through the activation of adenosine receptors (ARs) which belong to the G-protein-coupled receptors family and comprise four subtypes, namely A1, A2A, A2B, and A3 ARs. Activation of ARs modulates a variety of effector systems, although, classically, the adenosine-mediated signaling has been subdivided based

Adenosine Receptor Ligands as Potential Therapeutic Agents for. . .

91

on its ability to raise or lower cAMP levels. In fact, the A1 and A3 ARs preferentially couple to Gi/o proteins to inhibit adenylyl cyclase thus decreasing cAMP levels, whereas the A2A and A2B ARs stimulate adenylyl cyclase activity through activation of Gs proteins, thus increasing cAMP levels.

2 The Wound Healing Process Wounds repair process begins with the “hemostasis phase” that is characterized by vascular conscription and fibrin clot formation by platelets. Then, the clot and surrounding wound tissue release pro-inflammatory cytokines and growth factors such as transforming growth factor (TGF-β), platelet-derived growth factor (PDGF), and fibroblast growth factor (FGF). Once bleeding is stopped, inflammatory cells such as neutrophils, macrophages, and lymphocytes migrate into the wound thus promoting the “inflammatory phase” [1, 16]. Neutrophils are able, through phagocytosis activity, to both eliminate any bacteria and degrade necrotic tissue. Moreover, neutrophils release inflammatory mediators such as tumor necrosis factor (TNF-α), interleukins IL-1β and IL-6, and vascular endothelial growth factor (VEGF) which increases the vessels permeability and promotes the growth of new blood vessels [16, 17]. Macrophages play multiple roles in wound healing. In the early stage, they enter the lesion area and contribute to wound cleansing by phagocytosis. Then macrophages undergo a phenotypic transition to a reparative state that stimulates keratinocytes, fibroblasts, and the process of angiogenesis thus promoting tissue regeneration [18]. The “proliferative phase” of healing is characterized by a broad activation of fibroblasts, macrophages, and endothelial cells. Wound closure, matrix deposition, and angiogenesis take place during this phase. Fibroblasts are the main ones responsible for replacing the fibrin-rich platelet-derived matrix with granulation tissue. Fibroblasts, driven by signaling molecules including TFG-β and PDGF, degrade the provisional matrix and replace it with a granulation tissue rich in fibronectin, proteoglycans, and type III collagen, an immature form of collagen protein. This immature granulation tissue acts as a scaffold supporting both the formation of new blood vessels and the deposition of mature extracellular matrix (ECM) [19, 20]. Macrophages during this phase play a significant role in angiogenesis. New blood vessels are created to meet the metabolic demands of the highly proliferative healing tissue allowing nutrients and factors involved in the wound repair process to enter the injury site [21]. “Remodeling” is the last stage of wound healing and can last up to 2 years after injury. During this phase, the formation of granulation tissue stops due to apoptosis. Fibroblasts are the major cell type responsible for wound remodeling. At this stage, the components of ECM undergo some changes. Type III collagen, which was produced in the proliferative phase, is now replaced by type I collagen. Which

92

F. Varano et al.

increases the tensile strength of the forming scar. However, the integrity and architecture of scar ECM never fully return to that of unwounded tissue. Subsequently, myofibroblasts cause contractions of the wound and help to reduce the surface of the developing scar. In addition, the angiogenetic processes decrease, the blood flow of the wound is reduced, and the metabolic activity of the wound slows down and finally stops [19, 20]. It is important to underline that many factors, both endogenous and exogenous, can lead to impaired wound healing process. Among endogenous factors, aging is the most determinant causing a temporal delay in each phase of the process. Nutritional status is a crucial exogenous factor. In particular, a condition of malnutrition can compromise the activity of the immune system, causing healing disorders and infectious complications. Of course, the general health status of people can affect the speed and quality of healing. In particular, diabetic individuals, which total hundreds of millions around the world, are characterized by impaired healing of the skin wounds and are prone to developing chronic non-healing ulcers in the lower extremities known as diabetic foot ulcers [22].

3 Adenosine in Wound Healing During normal physiological conditions, the extracellular adenosine concentration is maintained low (30–300 nM) and constant. In contrast, under conditions of cellular or tissue necrosis, stress, or injury, its concentration rises dramatically to micromolar range, as a result of ATP catabolism. All the cells involved in the healing process, macrophages, epidermal cells, fibroblasts, and endothelial cells, express the various AR subtypes, although different cells and even the same cellular type, such as endothelial cells, express different AR subtypes [23, 24]. Adenosine is already involved in the early stages of the wound healing process, represented by the hemostatic and inflammatory phase. Numerous studies show that adenosine is a powerful regulator of the inflammatory process [25]. It has been observed that adenosine reduces inflammatory functions mainly through the activation of the A2A AR (Fig. 1). Studies with genetically modified animals showed that the absence of A2A ARs increased inflammatory damage [26, 27]. Adenosine acting at the A2A AR subtype can reduce the activation of neutrophils and macrophages, the differentiation and proliferation of T cells, the activity of NK cells and the production of pro-inflammatory cytokines. It has also been observed that, through activation of the A2A AR, adenosine induces the differentiation of macrophages into M2-type that promote wound healing through the release of factors such as VEGF. At the same time adenosine and A2A AR agonists strongly downregulate TNFα expression, thus promoting an angiogenic switch, shifting macrophages from an inflammatory to an angiogenic phenotype [28–30]. Furthermore, adenosine acting at all ARs has been recognized as a potent stimulator of angiogenesis which is a pivotal event in the wound healing process because injured tissue needs oxygen and nutrients to be efficiently repaired (Fig. 1).

Adenosine Receptor Ligands as Potential Therapeutic Agents for. . .

93

Fig. 1 Adenosine and ARs in wound healing and fibrosis

Studies in animal models showed that treatment with CGS 21680, a selective A2A AR agonist, promoted blood vessel formation and accelerated wound closure in wild-type mice, while having no effect on A2A AR knockout mice [31]. The principal pro-angiogenic actions of adenosine are due to its ability to control the production of pro-angiogenic substances from vascular and immune cells within the microenvironment of injured tissues. In fact, A1 AR mediated angiogenesis involves the release of VEGF from monocytes [32], while activation of the A2B subtype induces wound angiogenesis by upregulation in human microvascular endothelial cells of VEGF and endothelial NO synthase (eNOS) [33]. Adenosine, via the A2A and A2B ARs, stimulates angiogenesis by modulating thrombospondin-1 expression in human macrophages [34]. Moreover, adenosine through A2B and A3 ARs also promotes the production of VEFG, IL-8, and angiopoietin-1 from mast cells [35], and A1, A2A, and A2B AR selective agonists contribute to proliferation and wound healing in human EAhy926 endothelial cells [36]. Finally, activation of the A2A AR induces dermal fibroblast to produce collagen I and III, and to downregulate matrix metalloproteinases (MMP 9, 2, and 14) which are responsible for collagen disruption, thus evidencing a role of adenosine also in the tissue proliferation phase of the wound healing process. In fact, the blockade or deletion of this receptor subtype protects mice from bleomycin-induced dermal fibrosis which is a murine model of scleroderma [37, 38] (Fig. 1). In synthesis, adenosine receptor agonists, selective or not, may represent new and interesting therapeutic agents for the treatment of impaired wound healing which is one of the main concerns, especially for diabetic patients (up to 28% of diabetic foot ulcers can cause amputation). Topical application of A2A AR agonists accelerates the healing of skin wounds in healthy and diabetic rats, but not in A2A AR knockout mice [31, 39]. CGS21680 and sonedenoson (MRE-0094) (Fig. 2), two well-known A2A AR agonists, induce wound closure by a mechanism that depends on tissue plasminogen activator [40] and promote angiogenesis in wild-type mice. They heal wounds faster than beclapermin gel which is the only growth factor approved for

94

F. Varano et al.

Fig. 2 AR ligands evaluated in impaired wound healing and fibrosis

clinical use [31, 39]. Sonedenoson administered as a topical gel was tested as a potential drug for diabetic foot ulcers, but phase 2 clinical trial had poor enrollment and was terminated [41]. Despite this, encouraging data come from more recent studies which highlighted the tissue repair, anti-ischemic and anti-inflammatory properties of polydeoxyribonucleotide (PDRN) which is a proprietary and registered DNA-derived drug extracted from the sperm of trout bred for human food purposes. In vivo experiments demonstrated that PDRN promoted wound healing of diabetic foot ulcers better than placebo. All data obtained from these studies indicated that the most relevant mechanism of action of PDRN for promoting tissue repair was the A2A AR activation. In fact, DMPX (Fig. 2), a potent and selective A2A AR antagonist, was able to revert the PDRN-induced effects on wound healing. Indeed, it has been speculated that PDRN may represent a pro-drug able to generate active deoxyribonucleotides, nucleosides, and bases that interact with the A2A AR [42, 43].

Adenosine Receptor Ligands as Potential Therapeutic Agents for. . .

95

4 Adenosine and Fibrosis The observation that adenosine, acting at its receptors, stimulates the formation of ECM has suggested the possibility that excessive activation of ARs could lead to excess matrix production, scarring, and fibrosis of the skin and other organs, such as the liver, heart, lungs, and kidneys (Fig. 1). The A2A AR is expressed on human hepatic stellate cells, which are the fibroblasts of the liver, and its occupancy induces collagen I and III production by these cells. A2A AR knockout mice, unlike wild-type littermates, are protected from developing hepatic fibrosis following administration of the known hepatic fibrosisinducing agents CCl4 or thioacetamide, as well as wild-type mice treated with caffeine (Fig. 2), a nonselective AR antagonist, or with ZM231385 (Fig. 2) a highly selective A2A AR antagonist [44]. The pivotal role of adenosine and A2A AR in dermal fibrosis has been demonstrated in ADA deficient mice, a murine model in which both circulating and tissue adenosine levels are extremely high. Indeed, ADA is the key enzyme that catabolized adenosine into inosine. These genetically modified mice spontaneously develop dermal fibrosis which is diminished by treatment with an A2A AR antagonist [45]. Also, expression of A2B AR is significantly elevated in bleomycin-induced fibrosis, a well-established mouse model of dermal fibrosis, thus suggesting both a pro-fibrotic role for this receptor subtype too and a potential therapeutic use of A2B AR antagonists to treat dermal fibrosis [46]. Pharmacological blockade of A2A AR with ZM241385 decreased size and enhanced the tensile strength of the scar in a murine model that mimics human scarring. These data show that A2A AR antagonists can be used to reduce scarring, also improving the collagen composition of the healed wound [47]. Moreover, topical application of ZM241385 or A2A AR deletion prevents radiation-induced dermal injury in a murine model of irradiation-induced fibrosis. Therefore, A2A AR antagonism may be a useful strategy for preventing or ameliorating radiation dermatitis which is a common side effect of radiotherapy treatment of cancers [48]. Adenosine deaminase deficient mice also develop pulmonary inflammation and fibrosis that are significantly reduced by treatment with CVT-6883 (Fig. 2), a selective A2B AR antagonist. This result indicates that A2B AR signaling contributes to the pro-inflammatory and pro-fibrotic activities present in chronic lung diseases, such as severe asthma, COPD, and pulmonary fibrosis [49, 50]. On the contrary, adenosine A2B AR stimulation was shown to suppress collagen production in cardiac fibroblasts, suggesting a protective role of this receptor in myocardial fibrosis [51]. Reduction of cardiac fibrosis in a model of uremic cardiomyopathy is also obtained by the use of SVL320 (Fig. 2) a highly selective A1 AR antagonist [52]. The effects of ARs signaling in the development and progression of renal fibrosis indicate that, while A2A AR stimulation protects against inflammation, A3 and A2B AR activation promotes renal fibrosis. In fact, LJ-4459 (Fig. 2) [53], a dual-acting

96

F. Varano et al.

ligand that is an A2A AR agonist and an A3 AR antagonist, prevents the progression of tubule-interstitial fibrosis [54, 55]. From the above, the important role of adenosine in the pathophysiology of tissue fibrosis is evidenced. In fact, adenosine stimulates fibrosis in the skin, lungs, liver, and kidneys, while inhibiting myocardial fibrosis. Thus, adenosine receptor antagonism may represent a novel therapeutic strategy for the treatment of fibrotic diseases. A2A receptor antagonists could be used for the therapy of dermal and hepatic fibrosis, while A2B receptor blockade potentially leads to pulmonary fibrosis control.

5 Conclusions Adenosine plays important roles in inflammation, angiogenesis, and matrix production, which are critical for wound healing and tissue repair. There are a few FDA-approved therapies for the treatment of impaired healing which remains an unsolved clinical issue. Taking this into account, the development of AR agonists, either selective for the A2A receptor subtype or with a combined action at all the ARs, could lead to new drugs representing interesting alternative agents to beclapermin a growth factor derived drug, approved by the FDA for the treatment of chronic ulcers. In fact, the use of beclapermin has been drastically reduced due to its high production costs, its short storage stability along with an increased risk to malignant infections. On the other hand, AR agonists are in general small synthetic molecules of greater stability and lower cost compared to growth factors derived drugs. Moreover, adenosine plays a direct role in promoting fibrosis in the skin, lungs, and liver, whereas inhibiting cardiac fibrosis. Targeting of ARs to diminish fibrosis is currently under investigation, with the primary objective to find new therapies for these difficult clinical settings. Compliance with Ethical Standards Conflict of Interest: The Author declares that they have no conflict of interest. Funding: Original research of our team is funded by an intramural grant from the University of Florence (Fondi Ateneo Ricerca 2022). Ethical Approval: This chapter does not contain any studies with human participants or animals performed by the authors.

References 1. Rodrigues M, Kosaric N, Bonham CA, Gurtner GC (2019) Wound healing: a cellular perspective. Physiol Rev 99:665–706 2. Sen CK (2019) Human wounds and its burden an updated compendium of estimates. Adv Wound Care 8:39–48

Adenosine Receptor Ligands as Potential Therapeutic Agents for. . .

97

3. Nussbaum SR, Carter MJ, Fife CE, DaVanzo J, Haught R, Nusgart M et al (2018) An economic evaluation of the impact, cost, and medicare policy implication of chronic nonhealing wounds. Value Health 21:27–32 4. Armstrong DG, Wrobel J, Robbins JM (2007) Guest editorial: are diabetes-related wounds and amputations worse than cancer? Int Wound J 4:286–287 5. Smiell JM, Weiman J, Steed DL, Perry BH, Sampson AR, Schwab BH (1999) Efficacy and safety of becaplermin (recombinant human platelet-derived growth factor-BB) in patients with nonhealing, lower extremity diabetic ulcers: a combined analysis of four randomized studies. Wound Repair Regen 7:335–346 6. Zaulyanov L, Kirsner RS (2007) A review of a bi-layered living cell treatment (Apligraf) in the treatment of venous leg ulcers and diabetic foot ulcers. Clin Interv Aging 2:93–98 7. Hart CE, Loewen-Rodriguez A, Lessem J (2012) Dermagraft: use in the treatment of chronic wounds. Adv Wound Care 1:138–141 8. Wynn TA (2008) Cellular and molecular mechanisms of fibrosis. J Pathol 214:199–210 9. King TE, Pardo A, Selman M (2011) Idiopathic pulmonary fibrosis. Lancet 378:1949–1961 10. Gines P, Krag A, Abraldes JG, Sola E, Fabrellas N, Kamath PS (2021) Liver cirrhosis. Lancet 398:1359–1376 11. Hinderer S, Schenke-Layland K (2019) Cardiac fibrosis – a short review of causes and therapeutic strategies. Adv Drug Deliv Rev 146:77–82 12. Wang Z-C, Zhao W-Y, Cao Y, Liu Y-Q, Sun Q, Shi P, Cai J-Q, Shen XZ, Tan W-Q (2020) The roles of inflammation in keloid and hypertrophic scars. Front Immunol 11:603187 13. Straub JM, New J, Hamilton CD, Lomiska C, Shnayder Y, Thomass SM (2015) Radiationinduced fibrosis: mechanism and implications for therapy. J Cancer Res Clin Oncol 141:1985– 1994 14. Denton CP, Khanna D (2017) Systemic sclerosis. Lancet 390:1685–1699 15. Borea PA, Gessi S, Merighi S, Vincenzi F, Varani K (2018) Pharmacology of adenosine receptors: the state of the art. Physiol Rev 98:1591–1625 16. Chesko DM, Wilgus TA (2022) Immune cells in cutaneous wound healing: a review of functional data from animal models. Int J Mol Sci 23:2444 17. Werner S, Grose R (2003) Regulation of wound healing by growth factors and cytokines. Physiol Rev 83:835–870 18. Koh TJ, Dipietro LA (2011) Inflammation and wound healing: the role of the macrophage. Expert Rev Mol Med 13:E23 19. Xue M, Jackson CJ (2013) Extracellular matrix reorganization during wound healing and its impact on abnormal scarring. Adv Wound Care 4:119–136 20. Li B, Wang JHC (2011) Fibroblasts and myofibroblasts in wound healing: force generation and measurement. J Tissue Viability 20:108–120 21. Honnegowda TM, Kumar P, Udupa EG, Kumar S, Kumar U, Rao P (2015) Role of angiogenesis and angiogenic factors in acute and chronic wound healing. Plast Aesthet Res 2:243–249 22. Guo S, DiPietro LA (2010) Factors affecting wound healing. J Dent Res 89:219–229 23. Foektistov I, Biaggioni I, Cronstein BN (2009) Adenosine receptors in wound healing, fibrosis and angiogenesis. Handb Exp Pharmacol 193:383–397 24. Feoktisstov I, Goldstein AE, Ryzhov S, Zeng D, Belardinelli L, Voyno-Yasenetskaya T, Biaggioni I (2002) Differential expression of adenosine receptors in human endothelial cells. Role of A2B receptors in angiogenic factor regulation. Circ Res 90:531–538 25. Pasquini S, Contri C, Borea PA, Vincenzi F, Varani K (2021) Adenosine and inflammation: here, there and everywhere. Int J Mol Sci 22:7685 26. Haskò G, Pacher P (2008) A2A receptors in inflammation and injury: lessons learned from transgenic animals. J Leukoc Biol 83:447–455 27. Haskò G, Cronstein B (2013) Regulation of inflammation by adenosine. Front Immunol 4:85 28. Pinhal-Enfield G, Ramanathan M, Haskò G, Vogel SN, Salzman AL, Boons GJ, Leibovich SJ (2003) An angiogenic switch in macrophages involving synergy between toll-like receptors 2, 4, 7, and 9 and adenosine A2A receptors. Am J Pathol 163:711–721

98

F. Varano et al.

29. Macedo L, Pinhal-Enfield G, Alshits V, Elson G, Cronstein BN, Leibovich SJ (2007) Wound healing is impaired in MyD88-deficient mice. A role for MyD88 in the regulation of wound healing by adenosine A2A receptors. Am J Pathol 171:1774–1788 30. Leibovich SJ, Chen JF, Pinhal-Enfield G, Belem PC, Elson G, Rosania A, Ramanathan M, Montesinos C, Jacobson M, Schwarzschild MA, Fink JS, Cronstein B (2002) Synergistic up-regulation of vascular endothelial growth factor expression in murine macrophages by adenosine A2A receptor agonists and endotoxin. Am J Pathol 160:2231–2244 31. Montesinos MC, Desai A, Chen JF, Yee H, Schwarzschild MA, Fink JS, Cronstein BN (2002) Adenosine promotes wound healing and mediates angiogenesis in response to tissue injury via occupancy of A2A receptors. Am J Pathol 160:2009–2018 32. Auchampach JA (2007) Adenosine receptors and angiogenesis. Circ Res 101:1075–1077 33. Du X, Ou X, Song T, Zhang W, Cong F, Zhang S, Xiong Y (2015) Adenosine A2B receptor stimulates angiogenesis by inducing VEGF and eNOS in human microvascular endothelial cells. Exp Biol Med 240:1472–1479 34. Ernens I, Bousquenaud M, Lenoir B, Devaux Y, Wagner DR (2015) Adenosine stimulates angiogenesis by up-regulating production of trombospondin-1 by macrophages. J Leukoc Biol 97:9–18 35. Feoktistov I, Ryzhov S, Goldstein AE, Biaggioni I (2003) Mast cell-mediated stimulation of angiogenesis. Cooperative interaction between A2B and A3 adenosine receptors. Circ Res 92: 485–492 36. Bonyanian Z, Walker M, Du Toit E, Rose’Meyer RB (2019) Multiple adenosine receptor subtypes stimulate wound healing in human EA.hy926 endothelial cells. Purinergic Signal 15:357–366 37. Chan ESL, Fernandez P, Merchant AA, Montesinos MC, Trzaska S, Desai A, Tung CF, Khoa DN, Pillinger MH, Reiss AB, Tomic-Canic M, Chen JF, Schwarzschild MA, Cronstein BN (2006) Adenosine A2A receptors in diffuse dermal fibrosis. Pathogenic role in human dermal fibroblasts and in a murine model of scleroderma. Arthritis Rheum 54:2632–2642 38. Shaikh G, Zhang J, Perez-Aso M, Mediero A, Cronstein B (2016) Adenosine A2A receptor promotes collagen type III synthesis via β-catenin activation in human dermal fibroblasts. Br J Phamacol 173:3279–3291 39. Victor-Vega C, Desai A, Montesinos MC, Cronstein BN (2002) Adenosine A2A receptor agonists promote more rapid wound healing than recombinant human platelet-derived growth factor (Becaplermin gel). Inflammation 26:19–24 40. Montesinos MC, Desai-Merchant A, Cronstein BN (2015) Promotion of wound healing by agonist of adenosine A2A receptor is dependent on tissue plasminogen activator. Inflammation 38:2036–2041 41. Jacobson KA, Tosh DK, Jain S, Gao ZG (2019) Historical and current adenosine receptor agonists in preclinical and clinical development. Front Cell Neurosci 13:124 42. Squadrito F, Bitto A, Altavilla D, Arcoraci V, De Caridi G, De Feo ME, Corrao S, Pallio G, Sterrantino C, Minutoli L, Saitta A, Vaccaro M, Cucinotta D (2014) The effect of PDRN, an adenosine receptor A2A agonist, on the healing of chronic diabetic foot ulcers: results of a clinical trial. J Clin Endocrinol Metab 99:E746–E753 43. Galeano M, Pallio G, Irrera N, Mannino F, Bitto A, Altavilla D, Vaccaro M, Squadrito G, Arcoraci V, Colonna MR, Lauro R, Squadrito F (2021) Polydeoxyribonucleotide: a promising biological platform to accelerate impaired skin wound healing. Pharmaceuticals 14:1103 44. Chan ESL, Montesinos MC, Fernandez P, Desai A, Delano DL, Yee H, Reiss AB, Pillinger MH, Chen JF, Schwarzschild MA, Friedman SL, Cronstein BN (2006) Adenosine A2A receptors play a role in the pathogenesis of hepatic cirrhosis. Br J Pharmacol 148:1144–1155 45. Fernandez P, Trzaska S, Wilder T, Chiriboga L, Blackburn MR, Cronstein BN, Chan ESL (2008) Pharmacological blockade of A2A receptors prevents dermal fibrosis in a model of elevated tissue adenosine. Am J Pathol 172:1675–1682

Adenosine Receptor Ligands as Potential Therapeutic Agents for. . .

99

46. Karmouty-Quintana H, Molina JG, Philip K, Bellocchi C, Gudenkauf B, Wu M, Chen NY, Collum SD, Ko J, Agarwal SK, Assassi S, Zhong H, Blackburn MR, Weng T (2018) The antifibrotic effect of A2B adenosine receptor antagonism in a mouse model of dermal fibrosis. Arthritis Rheumatol 70:1673–1684 47. Perez-Aso M, Chiriboga L, Cronstein BN (2012) Pharmacological blockade of adenosine A2A receptors diminishes scarring. FASEB J 26:4254–4263 48. Perz-Aso M, Mediero A, Low YC, Levine J, Cronstein BN (2016) Adenosine A2A receptor plays an important role in radiation-induced dermal injury. FASEB J 30:457–465 49. Sun CX, Zhong H, Mohsenin A, Morschl E, Chunn JL, Molina BL, Zeng D, Blackburn MR (2006) Role of A2B adenosine receptor signaling in adenosine-dependent pulmonary inflammation and injury. J Clin Invest 116:2173–2182 50. Philip K, Weng Mills T, Davies J, Chen NY, Karmouty-Quintana H, Luo F, Molina JG, Amione-Guerra J, Sinha N, Guha A, Eltzschig HK, Blackburn MR (2017) HIF1A up-regulates the ADORA2B receptor on alternatively activated macrophages and contributes to pulmonary fibrosis. FASEB J 31:4745–4758 51. Wakeno M, Minamino T, Seguchi O, Okazaki H, Tsukamoto O, Okada K, HirataA FM, Asanuma H, Kim J, Komamura K, Takashima S, Mochizuki N, Kitakaze M (2006) Longterm stimulation of adenosine A2B receptors begun after myocardial infarction prevents cardiac remodeling in rats. Circulation 114:1923–1932 52. Kalk P, Eggert B, Relle K, Godes M, Heiden S, Sharkovska Y, Fischer Y, Ziegler D, Bielenberg GW, Hocher B (2007) The adenosine A1 receptor antagonist SVL320 reduces myocardial fibrosis in rats with 5/6 nephrectomy without affecting blood pressure. Br J Pharmacol 151: 1025–1032 53. Hou X, Majik MS, Kim K, Pyee Y, Alexander V, Chung HJ, Lee HW, Chandra G, Lee JH, Park S, Choi WJ, Kim HO, Phan K, Gao ZG, Jacobson KA, Choi S, Lee SK, Jeong LS (2012) Structure-activity relationships of truncated C2- or C8-substituted adenosine derivatives as dual acting A2A and A3 adenosine receptor ligands. J Med Chem 55:342–356 54. Pak ES, Jeong LS, Hou X, Tripathi SK, Lee J, Ha H (2021) Dual actions of A2A and A3 adenosine receptor ligand prevents obstruction-induced kidney fibrosis in mice. Int J Mol Sci 22:5667 55. Roberts VS, Cowan PJ, Alexander SI, Robson SC, Dwyer KM (2014) The role of adenosine receptors A2A and A2B signaling in renal fibrosis. Kidney Int 86:685–692

Top Med Chem (2023) 41: 101–142 https://doi.org/10.1007/7355_2023_162 © The Author(s), under exclusive license to Springer Nature Switzerland AG 2023 Published online: 19 August 2023

Adenosine A2A Receptor Antagonists: Chemistry, SARs, and Therapeutic Potential Andrea Spinaci, Michela Buccioni, Cui Chang, Diego Dal Ben, Beatrice Francucci, Catia Lambertucci, Rosaria Volpini, and Gabriella Marucci Contents 1 Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 2 A2A Adenosine Receptor Antagonists Evaluated in Clinical Trials . . . . . . . . . . . . . . . . . . . . . . . . 3 A2A Adenosine Receptor Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.1 Xanthine Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 3.2 Non-Xanthine Derivatives . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 4 Structural Features of A2A Structure and Its Interaction with Antagonists . . . . . . . . . . . . . . . . 5 Potential Application of A2A Adenosine Receptor Antagonists . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5.1 A2A Adenosine Receptor Antagonists and Neurodegenerative Disorders . . . . . . . . . . . 5.2 A2A Adenosine Receptor Antagonists and Neuroprotection . . . . . . . . . . . . . . . . . . . . . . . . . 5.3 A2A Adenosine Receptor Antagonists and Cancer . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 6 Concluding Remarks . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . .

102 103 105 105 107 120 124 124 126 127 129 130

Abstract Many physiological processes are modulated by the naturally occurred nucleoside adenosine after interaction with four G-protein-coupled receptor subtypes named A1, A2A, A2B, and A3. All adenosine receptors are widely distributed in the body. The A2A subtype is present in the central nervous system in the dopaminerich regions, in glutamatergic and GABAergic pathways intrinsic to the hippocampus, and in the cortex, where it is also co-expressed with dopamine D2 and cannabinoid CB1 receptors. The A2A adenosine receptor distribution also regards numerous peripheral tissues such as blood vessels, endothelial, lymphoid, smooth muscle cells, and neurons. Since it is an emerging receptor implicated in many pathologies and its blockade could be beneficial in neuroinflammation, neurodegenerative disorders, and cancer, this review focuses the attention on the chemical structures of selective A2A adenosine receptor antagonists, divided into different A. Spinaci, M. Buccioni, C. Chang, D. Dal Ben, B. Francucci, C. Lambertucci, R. Volpini (✉), and G. Marucci Medicinal Chemistry Unit, School of Pharmacy, University of Camerino, Camerino, MC, Italy e-mail: [email protected]

102

A. Spinaci et al.

chemical classes. One section is dedicated to the representation of published crystallographic structures and molecular modeling studies. Furthermore, the use of A2A adenosine receptor antagonists for the treatment of neurodegenerative disorders, as well as their potential as neuroprotective agents, will be discussed. Finally, due to the involvement of the A2A adenosine receptor in activated immune cells, it will be taken into consideration the potential application of A2A adenosine receptor antagonists in the anti-cancer therapy. Keywords A2A adenosine receptor antagonists, A2A adenosine receptor crystal structure, Cancer, Neurodegeneration, Neuroinflammation, Non-xanthine derivatives, Xanthine derivatives

1 Introduction Adenosine (Ado) is a natural nucleoside, which is widespread in mammalian organism tissues where it modulates a series of physiological processes through the interaction with four G-protein-coupled receptor subtypes named A1, A2A, A2B, and A3. Ado receptors (ARs) are distributed in all tissue and are often co-expressed in the same cell type [1]. In particular, the localization of A2AARs, by using pharmacological and immunohistochemical tools, was found, in the central nervous system (CNS), in the dopamine-rich regions, in glutamatergic and GABAergic pathways intrinsic to the hippocampus, and in the cortex [2]. In the CNS, A2AARs are co-expressed with dopamine D2 and cannabinoid CB1 receptors [3, 4].The distribution of A2AARs is not limited to the medium-sized spiny neurons in the basal ganglia. In fact, PCR and northern blot analysis demonstrated that the A2AAR gene is also expressed in numerous other tissues, namely blood vessels, endothelial cells, lymphoid cells, smooth muscle cells, and several neurons [5]. From its first cloning as an orphan receptor and deorphanization around the 1990s to today, at least 66 crystal and cryo-EM structures of A2AARs are known [6, 7], which allowed to depict the structural basis for the binding of agonists and antagonists. The therapeutic potential of this receptor ligands led to an intense medicinal chemistry efforts and many selective Ado receptor agonists and antagonists were discovered [8–10]. Commonly, the agonist structures are related to the natural ligand Ado while the antagonists belong to different chemical classes [11]. As the A2AAR is an emerging receptor implicated in many pathologies and its blockade could be useful in neuroinflammation [12, 13], neurodegenerative disorders [14, 15], and cancer [16, 17], this review draws the attention on the chemical structures of selective A2AAR antagonists. The high therapeutic potential underlying the modulation of the A2AAR has prompted researchers to design and develop increasingly potent and selective ligands to treat various clinical disorders. The first A2AAR antagonists resulted from the purines adenine and xanthine often combined with potentially reactive chemical moieties, such as the electron-rich ring furan and styrene group.

Adenosine A2A Receptor Antagonists: Chemistry, SARs, and. . .

103

Fig. 1 Structure and binding affinity of istradefylline (Ki nM)

The most important example is undoubtedly istradefylline (1, also called KW-6002, Fig. 1), which contains a xanthine ring and a 3,4-dimethoxystyryl substituent in the 8-position [18, 19]. It has been approved in Japan (2013), and then in the USA (2019), as a co-therapy for the treatment of Parkinson’s disease in association with levodopa/carbidopa to reduce off-time episodes and dyskinesias [20, 21]. Compounds with similar structure turned out to be poorly soluble in water and unstable, in a short time, due to photo-isomerization. In an attempt to get over these limitations, a large series of molecules possessing purine and non-purine heterocyclic scaffolds with considerable structural diversity were synthesized. Hence, the present review takes into consideration research on A2AAR antagonists from the first molecules possessing an alkyl xanthine structure to the more potent and selective heterocyclic compounds unrelated to xanthines. Then, a section will be dedicated at depicting the published crystallographic structures of the A2AARs and their interaction with antagonists. Furthermore, the use of Ado A2AAR antagonists for the treatment of neurodegenerative disorders, as well as their potential as neuroprotective agents, will be discussed. Finally, due to the involvement of the A2AAR in activated immune cells, it will be taken into consideration the potential application of A2AAR antagonists in the anti-cancer therapy.

2 A2A Adenosine Receptor Antagonists Evaluated in Clinical Trials Many A2AAR antagonists have been investigated in clinical trials for a variety of neurological diseases and cancers. A number of clinical trials regarding the natural occurring xanthine caffeine, one of the first AR antagonists discovered, have been already completed and some are still ongoing. At present, 1261 studies are present in the USA clinical trial portal (access to clinicaltrials.gov 26.10.2022) involving caffeine for its SNC psychostimulant and exercise and cognition effects, in the symptomatic Alzheimer’s disease even if its utility in some cases is debated. A comprehensive study report on caffeine has been recently published by Jacobson K.A. et al. [22]. Preladenant, also called SCH-420814 (2, Fig. 2) [23], reached the Phase III clinical trials for Parkinson disease (PD), but in contrast to istradefylline, it was discontinued due to lack of efficacy [24]. The A2AAR antagonist tozadenant (3, SYN115, Fig. 2) [10] entered in Phase III of clinical evaluation for Parkinson’s disease but was discontinued due to deaths from drug-induced agranulocytosis [21].

104

A. Spinaci et al.

Fig. 2 A2AAR antagonists evaluated in clinical trials. Binding affinity are in Ki nM and functional studies are in IC50 nM

The potent triazene A2AAR antagonist, AZD4365, also called HTL 1071 or imaradenant, (4, Fig. 2) [25] is in early clinical phase trial for cancer and it is showing encouraging results in enhancing antitumor immunity. Recently it was studied the encapsulation of AZD4635, with other active principles, into nanoparticles for intelligent drug delivery and the achievement of an excellent synergistic effect in photothermal therapy, chemotherapy, and immunotherapy [26]. The triazolopyrimidine vipadenant (5, BIIB014/V2006, Fig. 2) [27] was found efficacious in two phase II studies as an antiparkinsonian drug in association with levodopa and as monotherapy [28]; however, it was discontinued in June 2010 for toxicity problems (www.vernalis.com/media-centre/latest-releases/2010-releases/ 584). Another triazolopyrimidine ciforadenant (6, CPI444, Fig. 2) [29] is under

Adenosine A2A Receptor Antagonists: Chemistry, SARs, and. . .

105

Phase I/II trials in treatment-refractory renal cell cancer [30]. Etrumadenant (7, AB928, Fig. 2), an A2A/A2BAR mixed antagonist, is in clinical trials for cancer immunotherapy co-administered with doxorubicin. The strategy is based on the assumption that A2BAR activation leads to immunosuppressive effects in cancer microenvironment and preliminary studies showed promising results [31]. Taminadenant (8, PBF509/NIR178, Fig. 2) is in Phase I for the treatment of advanced non-small cell lung cancer (NSCLC). It is well tolerated in patients, causing controllable adverse effects, and produces an appreciable reduction in tumor mass [32]. The dual A2AAR antagonist/phosphodiesterase 10 (PDE-10) inhibitor PBF-999 (9, Fig. 2) has recently completed the phase I study for the treatment of solid tumors (ClinicalTrials.gov Identifier: NCT03786484). Other A2AAR antagonists are in phase I clinical trial studies to fight cancer or PD. One of these is inupadenant (10, EOS-850/EOS100850, Fig. 2; ClinicalTrials.gov Identifier: NCT03873883) which shows good human tolerability and initial evidence of clinical efficacy over solid tumors in patients not responding to standard treatment options [33].

3 A2A Adenosine Receptor Antagonists In the last decades, several heterocyclic classes of compounds, which can be divided into xanthine and non-xanthine derivatives, have been synthesized as A2AAR antagonists. As above mentioned, xanthines represent the prototypical group of antagonists, which have been modified to give a comprehensive collection of derivatives endowed with subtype selectivity. On the other hand, many other scaffolds, substituted in different positions and including tricyclic, bicyclic, and monocyclic derivatives, have been identified as selective A2AAR antagonists.

3.1

Xanthine Derivatives

As above mentioned, the xanthine caffeine and theophylline (11 and 12, Fig. 3) were the first A2AAR antagonists reported although with an affinity in the micromolar range and a low selectivity versus the other AR subtypes [9]. Theophylline is in therapy as anti-asthmatic drug while caffeine is used as an adjuvant in analgesic combinations. It was recently demonstrated that caffeine enhances the activity of non-steroidal analgesic drugs by pharmacodynamic interaction [34]. The 8-styrylxanthine analogue of caffeine, istradefylline (1, Fig. 1), was among the first A2AAR antagonists reported and the most representative compound of the xanthine family. As previously reported, 8-styrylxanthines are photosensitive and in solution the styryl group isomerizes from trans to cis conformation with consequent loss of activity. The simultaneous substitution in the 1,3,7 positions of the 8-styryl xanthine nucleus has given rise to interesting compounds, among which the

106

A. Spinaci et al.

Fig. 3 Structures and binding affinity of xanthine derivatives (Ki nM)

(E )-8-(3,4-dimethoxystyryl)-1,3-dipropyl-7-methylxanthine (13, (E )-KF17837; KiA2A = 1 nM at rat receptor; selectivity A1/A2A = 62, Fig. 3) resulted in one of the first A2A antagonists showing high selectivity versus the A1 subtype [35, 36]. Other A2A receptor-selective 8-styrylxanthines extensively studied comprise CSC (14, 8-(3-chlorostyryl)caffeine; Ki = 54 nM on rat receptor) [37] and its congener MSX-2 (15, 3-(3-hydroxypropyl)-8-(m-methoxystyryl)-1propargylxanthine) (Ki = 8 nM and 5 nM on rat brain striatal membranes and human recombinant A2AAR, respectively). Interestingly CSC and its derivatives resulted to be inhibitors of monoamine oxidase-B (MAO-B) in addition to A2AAR [38, 39]. The dual mechanism of action makes these drugs particularly suitable for the treatment of PD [2]. In fact, since MAO-B appears to be one of the main catabolic pathways of dopamine in the striatum, the block of this enzyme slows down the elimination of dopamine and increases its stay in the synapse. On the other hand, A2AAR blockade causes an increase in dopamine affinity versus the D2 dopamine receptor and clinical studies revealed that A2AAR antagonists improve motor dysfunctions of PD by reducing side effects such as dyskinesia. MSX2 belongs to the series of propargylxanthines, an extensively studied class of A2AAR antagonists, among which the 3,7-dimethyl-1-propargylxanthine (16, DMPX) is considered the first synthetic A2AAR antagonist endowed with xanthine core even if it showed low affinity and selectivity [40]. High selective derivatives of DMPX were developed by Muller et al. Among these, particular attention deserves the compounds 8-(m-bromostyryl)-3,7-dimethyl1-propargylxanthine (17, BS-DMPX) and 8-(m-methoxystyryl)-3,7-dimethyl-1-

Adenosine A2A Receptor Antagonists: Chemistry, SARs, and. . .

107

propargylxanthine (18, CS-DMPX) which, like the lead compound, have a poor water solubility. A useful approach to increase water solubility was the preparation of phosphate or amino acid prodrugs, in which the polar group/moiety is cleaved off in vivo to release the active compound [22, 41–43]. In particular, from MSX-2 derived two interesting compounds, the phosphate prodrug MSX-3, 19, and the L-valine ester prodrug MSX-4, 20. Binding studies confirmed that MSX-3 is readily cleaved by phosphatases to MSX-2, showing a Ki value equal to that obtained for its parent compound (KiA2A = 8 nM, on rat brain striatal membranes). Both MSX-3 and MSX-4 are commonly used tools in research, particularly in in vivo studies [44– 46].

3.2 3.2.1

Non-Xanthine Derivatives Triazolo-Quinazoline and -Quinoxaline Derivatives

First examples of heterocycles derivatives different from xanthines were the triazoloquinazoline CGS15943 (21), which was discovered in a search for benzodiazepine receptor modulators [47, 48], and the triazoloquinoxaline CP-66,713 (23) [49] and their congeners 22 and 24 (Fig. 4). These tricyclic compounds were found to possess very high A2AAR affinity at rat and human receptors with Ki or IC50 in the nM and sub nM range, although they resulted poorly selective. About 10 years later, a series of triazoloquinoxaline-1-one derivatives were reported; among them, the 4-amino-6-(benzylamino)-2-phenyl-[1,2,4]triazolo [4,3-a]quinoxalin-1(2H )-one (25) was found to possess high A2AAR affinity and selectivity. Removal of the N-benzyl group shifted the selectivity versus the A1AR receptor [50]. A subsequent study on 1,2,4-triazolo[4,3-a]quinoxalin-1-one derivatives demonstrated that replacement of the benzylamino group phenyl ring with other heterocycles like furan, thiophene, or pyridine led to compounds which maintained high A2AAR affinity although resulted less selective especially versus the A1AR [51].

Fig. 4 Structures and binding affinity of triazoloquinazoline and triazoloquinoxalines (Ki or IC50 nM)

108

3.2.2

A. Spinaci et al.

Pyrazolo-, Imidazolo-, and Thiazolo-Triazolopyrimidine Derivatives

Isosteric substitution of CGS15943 phenyl ring with a pyrazole nucleus led to A2AAR antagonists which maintained high affinity but were rather unselective. An example is represented by 8FB-PTP (26; Fig. 5), which opens the way to the search for new A2AAR antagonists with pyrazolotriazolopyrimidine scaffold [52]. Then, a number of potent and selective compounds were synthesized allowing to depict structure–activity relationships (SAR), which revealed the importance of the free amino group at the 5-position and the furane ring in the 2-position for the affinity and selectivity. Furthermore, substitution of the pyrazole ring in 7-position was more favorable for the A2A selectivity than that in 8-position as well as the presence of the pyrazole ring with respect to the triazole moiety [10, 53]. In particular, the presence at the 7-position of the pyrazole nucleus of a phenylalkyl chain improved the A2A affinity and selectivity, especially when an amino, hydroxy, and a methoxy group substituted the phenyl para position. These compounds 27-31 were found among the most potent and selective A2AAR antagonists with affinity in the nM and sub-nM range (Fig. 5). In particular, the 2-(furan-2-yl)-7-(3-(4-methoxyphenyl)propyl)-7Hpyrazolo[4,3-e][1,2,4]triazolo[1,5-c]pyrimidin-5-amine (30, SCH-442416, KiA2A = 0.048 nM) results endowed with the highest affinity and a selectivity of about 23,000-fold versus the A1 subtype and greater than 200,000-fold versus the other human AR subtypes. Later report did not confirm this extremely high affinity, which however remained in the low nM range [54]. Unfortunately, attempts to increase the water solubility of these molecules by replacement of the phenyl para substituent with a carboxylic or a more polar sulfonic group decreased the A2A binding affinity [55]. Subsequent modifications of these compounds led to the discovery of a series of pyrazolotriazolopyrimidines bearing at the 7-position of the pyrazole ring various arylpiperazine groups. Among them, the orally active

Fig. 5 Structures and binding affinity of pyrazolo- and imidazolo-triazolopyrimidines (Ki, nM)

Adenosine A2A Receptor Antagonists: Chemistry, SARs, and. . .

109

Fig. 6 Structure and binding affinity of thiazolotriazolopyrimidines (Ki nM)

methoxyethoxy ether derivative, preladenant (2, SCH-420814, Fig. 2), showed high affinity at both rat and human A2AAR with Ki of 2.5 nM and 1.1 nM, respectively, and very good selectivity versus the other AR subtypes and a panel of 59 receptors, enzymes, and ion channels together with favorable pharmacokinetic properties in rats. These characteristics make it a widely studied A2AAR antagonists in in vivo animal studies and clinical trials [23, 56]. Recently, Iteos Therapeutics patented a series of thiazolotriazolopyrimidines, bearing a carbonyl group in the thiazole ring, which have been designed as new A2AAR antagonists with low CNS penetrance and potentially useful in the treatment of peripheral cancers. Among them, inupadenant (10, Fig. 2) showed very high activity at A2AAR with an IC50 of 0.6 nM in the inhibition of cAMP production [57]. A novel series of [1,2,4]triazolo[5,1-f]purin-2-one derivatives by Advinus Therapeutics Ltd. were reported to display functional antagonism at the A2AAR with good selectivity over other AR subtypes. Among reported compounds, 32 (Fig. 5) was the most active with a Ki of 1.5 nM at human A2AARs measured in binding experiments. This compound showed good in vitro and in vivo pharmacokinetics properties and demonstrated good effectiveness in two models of Parkinson’s disease (haloperidol-induced catalepsy and 6-OHDA lesioned rat models) and depression (tail suspension and forced swim tests mice models) [58]. The design, synthesis, and in silico molecular docking of new thiazolotriazolopyrimidine derivatives as potent and selective A2AAR antagonists was undertaken by Mishra et al. [59]. In silico molecular docking studies revealed that all compounds exhibited strong interaction with A2AAR, which was confirmed by binding studies at human A2AARs stably expressed in HEK293 cells [59]. In fact, the thiazolotriazolopyrimidine derivatives 33 and 34 (KiA2A = 0.016 nM and 0.0082 nM, respectively, Fig. 6) demonstrated very high binding affinity and selectivity for the A2AAR. 33 was able to counteract the haloperidol-induced hypo-locomotor behavior (catalepsy and akinesia) in PD animal model producing motor stimulant effects. The synthesis and biological evaluation of 8-(furan-2-yl)-3-phenethylthiazolo [5,4-e] [1,2,4] triazolo[1,5c]pyrimidine-2(3H )-thione (35, PTTP, Fig. 6) were performed by Kumari et al. [60]. This compound showed a good affinity for A2AAR (KiA2A = 6.3 nM) and selectivity versus the A1AR (A1/A2A = 4,603). Docking analysis revealed that PTTP and SCH-58261 shared almost similar binding motif inside the transmembrane regions and extracellular loops (ECL2) at the human A2AAR co-crystallized with ZM241385. cAMP functional study, in HEK293T, cells demonstrated that this compound is an antagonist with a potency better than Scheme

110

A. Spinaci et al.

58261. In addition, in PD animal model, the pre-treatment with 35 attenuated catalepsy and akinesia without significant neurotoxicity in rotarod test.

3.2.3

Triazolo-Triazine, -Pyrimidine, and -Pyrazine Derivatives

The tricyclic scaffold of the triazoloquinazolines was simplified by researchers of the Zeneca group, which synthesized a series of bicyclic compounds including the triazolotriazine 4-(2-((7-amino-2-(furan-2-yl)-[1,2,4]triazolo[1,5-a][1,3,5]triazin-5yl)amino)ethyl)phenol (36, ZM 241385, Fig. 7), which showed high A2AAR affinity and good selectivity (KiA2A = 1.78 nM and 16.6 nM at rat and human receptor, respectively) [61, 62]. Indeed, later studies demonstrated ZM241385 to be an inverse agonist at the A2AAR [63]. This compound, which resulted in one of the most potent A2AAR ligands reported, showed a good activity also at the A2BAR subtype and has rapidly become the reference compound for the study of the A2AAR subtype, also in the tritiated form [64]. A number of modifications of the ZM241385 phydroxyphenylethylamino chain were reported but none of the resulted compounds showed higher affinity [65]. Substitution of the furyl ring with an ethoxy group decreased the A2AAR affinity of about 100 times (37, KiA2A = 178 nM), while removal of the 2-amino chain was detrimental for the A2AAR affinity (38, KiA2A = 2,029 nM) [66]. In 2009, researchers of Vernalis (R&D) Ltd. reported the synthesis of a series of functionalized triazolo[4,5-d]pyrimidine derivatives as A2AAR antagonists. Among them, vipadenant (5, BIIB014/V2006, Fig. 2) showed to possess high A2AAR affinity (Ki = 1.3 nM) and good selectivity versus the other AR subtypes, especially A3ARs [27]. Furthermore, this compound demonstrated very good

Fig. 7 Structures of triazolotriazines and triazolopyrimidines. Binding affinity are in Ki nM

Adenosine A2A Receptor Antagonists: Chemistry, SARs, and. . .

111

preclinical pharmacokinetic properties and was found orally active and very potent in animal models of Parkinson’s disease [67]. These characteristics allowed clinical evaluation of the compound in collaboration with Biogen Idec but, as already mentioned, despite positive clinical results, it was discontinued for toxicity problems. Modification of the vipadenant benzylic chain led to the analogue ciforadenant (6, CPI-444, Fig. 2), bearing at the 3-position of the triazolopyrimidine nucleus a substituted pyridine [68]. This compound showed high A2AAR affinity (Ki = 3.5 nM) and a better selectivity versus the A2B subtype (more than 400 times) with respect to vipadenant and was able, through the A2AAR blockade, to neutralize the Ado signaling and to enable antitumor immunity in preclinical models of cancers [29]. Recently, a number of 8-amino-6-aryl-1,2,4-triazolo[4,3-a]pyrazin-3-ones derivatives were reported to possess very high affinity for the human A2AAR combined, in some cases, with an extremely high selectivity. Compound 39 (Fig. 7) being the most relevant of the first series with a Ki = 2.9 nM. It was also tested to evaluate its ability to inhibit cAMP production with an IC50 of 98 nM [69]. Successively, the same research group reported the synthesis of other derivatives of the same scaffold and demonstrated that, on the basis of substituents present in 2- and 6-positions, it was possible to obtain potent and dual A1/A2AAR ligands with Ki values in the low nM range on both subtypes (40 and 41, Fig. 7). In a neuroprotection assay, the compounds were evaluated for their ability in counteracting β-amyloid peptide (Aβ)-induced toxicity on SH-SY5Y cells. Both of them significantly prevented toxicity induced by Aβ peptide or restored viability to control level highlighting their potential as new neuroprotective agents in Alzheimer’s disease (AD) [70]. The introduction of a functionalized alkyl-amino chain at the phenyl in 6-position furnished the 3-amino-N-(4-(8-amino-3-oxo-2phenyl-2,3-dihydro-[1,2,4]triazolo[4,3-a]pyrazin-6-yl)phenyl)propenamide (42), which exhibited a Ki = 0.59 nM and a selectivity more than 800-fold versus all the other ARs [71]. Its bromo derivative 43 (KiA2A = 10.6 nM, Fig. 7) was very efficacious as neuroprotective agent in the in vitro model of AD previously described [70].

3.2.4

Thiazolo- and Thieno-Pyrimidine Derivatives

A series of N5-substituted-2-(2-furanyl)thiazolo[5,4-d]pyrimidine-5,7-diamine derivatives was synthesized and pharmacologically characterized by Varano et al. [72] The introduction of a methoxy substituent at the 2- or 3-position of the phenyl ring of N5-benzyl-2-(2-furanyl)[1,3]thiazolo[5,4-d]pyridine-5,7-diamine (44, KiA2A = 22 nM, KiA1 = 43 nM, and KiA3 = 37 nM; Fig. 8), which is an unselective compound with nM affinity, led to derivatives 2-(2-furanyl)-N5-(2-methoxybenzyl) [1,3]thiazolo[5,4-d]pyrimidine-5,7-diamine (45, Fig. 8) and 2-(2-furanyl)-N5(3-methoxybenzyl)[1,3]thiazolo[5,4-d]pyrimidine-5,7-diamine (46, Fig. 8), respectively. These two compounds showed a peculiar behavior with two different affinity values for the A2AAR; a high affinity Ki value in the femtomolar range (KiH = 3.55

112

A. Spinaci et al.

Fig. 8 Structure and binding affinity of thiazolopyrimidines (Ki nM; KiH, fM). * KiHA2A and KiLA2A are referred to high affinity and low affinity calculated in a biphasic response curve obtained in binding studies

and 5.31 fM, respectively) and a low affinity Ki value in the nanomolar order (KiL = 6.45 and 26 nM, respectively). Moreover, 45 and 46 were able to inhibit cAMP in the basal condition behaving as inverse agonists (IC50 = 1.9 and 8.3 pM, respectively). The same authors synthesized similar analogues bearing different substituents at 2- and 5- position of the thiazolopyrimidine core [73]. Generally, these compounds showed high affinity toward the A1AR and A2AAR behaving as dual A1AR/A2AAR antagonists/inverse agonists. The N5-(2-methoxybenzyl)-2-phenyl-thiazolo[5,4-d]pyrimidine-5,7-diamine (47, Fig. 8) presented the highest affinity (KiA1 = 10.2 nM; KiA2A = 4.72 nM) and behaved as a potent A1/A2A antagonist/ inverse agonist (IC50A1 = 13.4 nM; IC50A2A = 5.34 nM). Then, other 7-aminothiazolo[5,4-d]pyrimidines, bearing different substituents at 2-, 5-, and 7-position of the thiazolopyrimidine scaffold, were designed, synthesized, and biologically evaluated [74]. Results showed that affinity and selectivity of the 7-amino-thiazolo [5,4-d]pyrimidine derivatives toward the AR subtypes can be modulated by the nature of the groups linked at the different positions of the bicyclic scaffold. The 5-(furan-2-yl)-2-phenylthiazolo[5,4-d]pyrimidin-7-amine (48, KiA2A = 1.7 nM Fig. 8) showed an nM affinity. Furthermore, also sub-nM binding affinities were found in compounds with a 7-amino-2-arylmethyl-thiazolo[5,4-d]pyrimidine structure [75]. The 2-(2-fluorobenzyl)-5-(furan-2yl)-thiazolo[5,4-d]pyrimidin-7-amine (49, KiA1 = 1.9 nM; KiA2A = 0.06 nM; Fig. 8) was evaluated for its antidepressant-like activity in in vivo studies in mice, showing an effect comparable to that of the reference compound amitriptyline. The synthesis of molecules bearing thieno[2,3-d]pyrimidine scaffold led to compounds with weak affinity toward A1ARs, but good binding affinity at A2A and A3ARs [76]. Compound showing the best A2AAR affinity was the 5,6-dimethyl-2-(benzamide)thieno[2,3-d]pyrimidin-4 (3H )-one (50, KiA1 > 30 μM, KiA2A = 650 nM, and KiA3 = 124 nM; Fig. 9). Compounds bearing the thieno[2,3-d]pyrimidine scaffold, substituted at the 6-position with various aryl and heteroaryl moieties, were also reported by Shook

Adenosine A2A Receptor Antagonists: Chemistry, SARs, and. . .

113

Fig. 9 Structures and binding affinity of thienopyrimidines (Ki nM)

Fig. 10 Structure and binding affinity of benzothiazole and pyrimidothiazole derivatives (Ki nM)

et al. and proved to possess enhanced A2AAR affinity [77]. These compounds were potent A2A antagonists having good in vivo efficacy in the mouse catalepsy model. Unfortunately, they showed a short half-life, limiting their use. Among these compounds are the chlorinebenzylthieno[2,3-d]pyrimidine derivatives, which showed good in vitro potency when a methylfuran (51, Fig. 9) and a 3-cyanophenyl (52, Fig. 9) substituted the scaffold 6-position.

3.2.5

Benzo-, Pyrimido-, and Oxazolo-Thiazolo Derivatives

The best of this class of molecules is tozadenant (3, SYN-115, Fig. 2), having a benzothiazole- based scaffold, which was identified by Hoffmann-La Roche [78]. It is a potent and selective A2AAR antagonist with good ADME properties; nevertheless, as reported above, even if entered in Phase III of clinical evaluation for PD, it was discontinued due to deaths from drug-induced agranulocytosis. On this basis, Basu et al. reported the design, synthesis, and biological evaluation of novel series of 2-substituted benzothiazole and thiazolo[5,4-c]pyridine derivatives aimed to discover novel, potent, and selective A2AAR antagonists with desirable physicochemical, pharmacokinetic, and pharmacological properties [79]. The results of the binding assay allowed to select three compounds as potent and selective A2AAR antagonists (53-55, Fig. 10). These three compounds were also assessed for their stability in liver microsomes. They did not show any CYP liability against major CYPs (